Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 1571, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383567

RESUMEN

Astrocytes express ionotropic receptors, including N-methyl-D-aspartate receptors (NMDARs). However, the contribution of NMDARs to astrocyte-neuron interactions, particularly in vivo, has not been elucidated. Here we show that a knockdown approach to selectively reduce NMDARs in mouse cortical astrocytes decreases astrocyte Ca2+ transients evoked by sensory stimulation. Astrocyte NMDAR knockdown also impairs nearby neuronal circuits by elevating spontaneous neuron activity and limiting neuronal recruitment, synchronization, and adaptation during sensory stimulation. Furthermore, this compromises the optimal processing of sensory information since the sensory acuity of the mice is reduced during a whisker-dependent tactile discrimination task. Lastly, we rescue the effects of astrocyte NMDAR knockdown on neurons and improve the tactile acuity of the animal by supplying exogenous ATP. Overall, our findings show that astrocytes can respond to nearby neuronal activity via their NMDAR, and that these receptors are an important component for purinergic signaling that regulate astrocyte-neuron interactions and cortical sensory discrimination in vivo.


Asunto(s)
Astrocitos , Receptores de N-Metil-D-Aspartato , Ratones , Animales , Astrocitos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Vibrisas/metabolismo , Neuronas/metabolismo , Transducción de Señal
2.
Animals (Basel) ; 11(8)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34438886

RESUMEN

Animal welfare labeling schemes have been developed to respond to consumers' expectations regarding farm animal welfare. They are designed to certify that labeled products comply with certain animal welfare standards. In this study, 12 pig welfare labeling schemes were reviewed, and their criteria related to pig welfare were compared. Information regarding farrowing criteria, space allowance, outdoor access, mutilations, and provision of enrichments and bedding material were gathered from the labels' internet pages and documentation. The results indicated a substantial variation between the labels in terms of the level of animal welfare they ensure. While certain schemes barely exceeded the minimum standards for the protection of pigs in the European Union, more demanding tiers of the multitier schemes had the potential to improve animal welfare substantially. The most ambitious tiers of multistage schemes were often comparable to organic standards providing outdoor facilities and additional space. The heterogeneity of the labels' standards complicates the comparison of labels.

3.
Neuropharmacology ; 187: 108502, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33631119

RESUMEN

There is emerging interest in the role of poly(ADP-ribose) polymerase-1 (PARP-1) in neurodegeneration and potential of its therapeutic targeting in neurodegenerative disorders. New generations of PARP inhibitors exhibit polypharmacological properties; they do not only block enzymatic activity with lower doses, but also alter how PARP-1 interacts with DNA. While these new inhibitors have proven useful in cancer therapy due to their ability to kill cancer cell, their use in neurodegenerative disorders has an opposite goal: cell protection. We hypothesize that newer generation PARP-1 inhibitors jeopardize the viability of dividing CNS cells by promoting DNA damage upon the PARP-DNA interaction. Using enriched murine astrocyte cultures, our study evaluates the effects of a variety of drugs known to inhibit PARP; talazoparib, olaparib, PJ34 and minocycline. Despite similar PARP enzymatic inhibiting activities, we show here that these drugs result in varied cell viability. Talazoparib and olaparib reduce astrocyte growth in a dose-dependent manner, while astrocytes remain unaffected by PJ34 and minocycline. Similarly, PJ34 and minocycline do not jeopardize DNA integrity, while treatment with talazoparib and olaparib promote DNA damage. These two drugs impact astrocytes similarly in basal conditions and upon nitrosative stress, a pathological condition typical for neurodegeneration. Mechanistic assessment revealed that talazoparib and olaparib promote PARP trapping onto DNA in a dose-dependent manner, while PJ34 and minocycline do not induce PARP-DNA trapping. This study provides unique insight into the selective use of PARP inhibitors to treat neurodegenerative disorders whereby inhibition of PARP enzymatic activity must occur without deleteriously trapping PARP onto DNA.


Asunto(s)
Astrocitos/efectos de los fármacos , Daño del ADN/efectos de los fármacos , ADN/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Astrocitos/metabolismo , Supervivencia Celular/efectos de los fármacos , Ratones , Minociclina/farmacología , Enfermedades Neurodegenerativas , Fenantrenos/farmacología , Ftalazinas/farmacología , Piperazinas/farmacología
4.
Cancers (Basel) ; 12(11)2020 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-33138032

RESUMEN

PTEN mutation occurs in a variety of aggressive cancers and is associated with poor patient outcomes. Recent studies have linked mutational loss of PTEN to reduced RAD51 expression and function, a key factor involved in the homologous recombination (HR) pathway. However, these studies remain controversial, as they fail to establish a definitive causal link to RAD51 expression that is PTEN-dependent, while other studies have not been able to recapitulate the relationship between the PTEN expression and the RAD51/HR function. Resolution of this apparent conundrum is essential due to the clinically-significant implication that PTEN-deficient tumors may be sensitive to poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) commonly used in the clinical management of BRCA-mutated and other HR-deficient (HRD) tumors. METHODS: Primary Pten-deficient (and corresponding wild-type) mouse embryonic fibroblasts (MEFs) and astrocytes and PTEN-null human tumor cell lines and primary cells were assessed for RAD51 expression (via the Western blot analysis) and DNA damage repair analyses (via alkali comet and γH2AX foci assays). RAD51 foci analysis was used to measure HR-dependent DNA repair. Xrcc2-deficient MEFs served as an HR-deficient control, while the stable knockdown of RAD51 (shRAD51) served to control for the relative RAD51/HR-mediated repair and the phospho-53BP1 foci analysis served to confirm and measure non-homologous end joining (NHEJ) activity in PTEN-deficient and shRAD51-expressing (HRD) lines. Cell proliferation studies were used to measure any potential added sensitivity of PTEN-null cells to the clinically-relevant PARPi, olaparib. RAD51 levels and DNA damage response signaling were assessed in PTEN-mutant brain tumor initiating cells (BTICs) derived from primary and recurrent glioblastoma multiforme (GBM) patients, while expression of RAD51 and its paralogs were examined as a function of the PTEN status in the RNA expression datasets isolated from primary GBM tumor specimens and BTICs. RESULTS: Pten knockout primary murine cells display unaltered RAD51 expression, endogenous and DNA strand break-induced RAD51 foci and robust DNA repair activity. Defective HR was only observed in the cells lacking Xrcc2. Likewise, human glioblastoma multiforme (GBM) cell lines with known PTEN deficiency (U87, PTEN-mutated; U251 and U373, PTEN-null) show apparent expression of RAD51 and display efficient DNA repair activity. Only GBM cells stably expressing shRNAs against RAD51 (shRAD51) display dysfunctional DNA repair activity and reduced proliferative capacity, which is exacerbated by PARPi treatment. Furthermore, GBM patient-derived BTICs displayed robust RAD51 expression and intact DNA damage response signaling in spite of PTEN-inactivating mutations. RNA expression analysis of primary GBM tissue specimens and BTICs demonstrate stable levels of RAD51 and its paralogs (RAD51B, RAD51C, RAD51D, XRCC2, XRCC3, and DMC1), regardless of the PTEN mutational status. CONCLUSIONS: Our findings demonstrate definitively that PTEN loss does not alter the RAD51 expression, its paralogs, or the HR activity. Furthermore, deficiency in PTEN alone is not sufficient to impart enhanced sensitivity to PARPi associated with HRD. This study is the first to unequivocally demonstrate that PTEN deficiency is not linked to the RAD51 expression or the HR activity amongst primary neural and non-neural Pten-null cells, PTEN-deficient tumor cell lines, and primary PTEN-mutant GBM patient-derived tissue specimens and BTICs.

5.
Glia ; 68(7): 1421-1434, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32036619

RESUMEN

Chronic neuroinflammation driven by microglia is a characteristic feature associated with numerous neurodegenerative diseases. While acute inflammation can assist with recovery and repair, prolonged microglial pro-inflammatory responses are known to exacerbate neurodegenerative processes. Yet, detrimental outcomes of extended microglial activation are counterbalanced by beneficial outcomes including phagocytosis and release of trophic factors promoting neuronal viability. Our past work has shown that the nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) is a key signaling hub driving pro-inflammatory microglia responses, but the signaling pathway maintaining PARP-1 activation remains elusive. While best understood for its role in promoting DNA repair, our group has shown that PARP-1 activity can be stimulated via Ca2+ influx-dependent ERK1/2-mediated phosphorylation. However, to date, the route of Ca2+ entry responsible for stimulating PARP-1 has not been identified. A likely candidate is via Ca2+ -permeable transient receptor potential melastatin 2 (TRPM2) channels activated downstream of PARP-1 in a cascade that involves ADP-ribose (ADPR) production by poly(ADP-ribose) glycohydrolase (PARG). Here we demonstrate that NMDA receptor (NMDAR) stimulation in primary cultured microglia induces their proliferation, morphological activation and release of pro-inflammatory mediators. These responses were contingent on the recruitment of PARP-1, PARG and Ca2+ permeable TRPM2 channels. Furthermore, we show that Ca2+ influx is necessary to activate PARP-1/TRPM2 signaling, in an ERK1/2-dependent, but DNA damage independent, manner. Our findings, showing that PARP-1/TRPM2 mediate the pro-inflammatory effects of NMDAR stimulation, provides a unifying mechanism linking elevated glutamate levels to chronic neuroinflammation.


Asunto(s)
Muerte Celular/fisiología , Inflamación/metabolismo , Transporte Iónico/fisiología , Microglía/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Peróxido de Hidrógeno/metabolismo , Ratones , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPM/metabolismo
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3353-3367, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30055293

RESUMEN

Cardiolipin (CL) is a key mitochondrial phospholipid essential for mitochondrial energy production. CL is remodeled from monolysocardiolipin (MLCL) by the enzyme tafazzin (TAZ). Loss-of-function mutations in the gene which encodes TAZ results in a rare X-linked disorder called Barth Syndrome (BTHS). The mutated TAZ is unable to maintain the physiological CL:MLCL ratio, thus reducing CL levels and affecting mitochondrial function. BTHS is best known as a cardiac disease, but has been acknowledged as a multi-syndrome disorder, including cognitive deficits. Since reduced CL levels has also been reported in numerous neurodegenerative disorders, we examined how TAZ-deficiency impacts cognitive abilities, brain mitochondrial respiration and the function of hippocampal neurons and glia in TAZ knockdown (TAZ kd) mice. We have identified for the first time the profile of changes that occur in brain phospholipid content and composition of TAZ kd mice. The brain of TAZ kd mice exhibited reduced TAZ protein expression, reduced total CL levels and a 19-fold accumulation of MLCL compared to wild-type littermate controls. TAZ kd brain exhibited a markedly distinct profile of CL and MLCL molecular species. In mitochondria, the activity of complex I was significantly elevated in the monomeric and supercomplex forms with TAZ-deficiency. This corresponded with elevated mitochondrial state I respiration and attenuated spare capacity. Furthermore, the production of reactive oxygen species was significantly elevated in TAZ kd brain mitochondria. While motor function remained normal in TAZ kd mice, they showed significant memory deficiency based on novel object recognition test. These results correlated with reduced synaptophysin protein levels and derangement of the neuronal CA1 layer in hippocampus. Finally, TAZ kd mice had elevated activation of brain immune cells, microglia compared to littermate controls. Collectively, our findings demonstrate that TAZ-mediated remodeling of CL contributes significantly to the expansive distribution of CL molecular species in the brain, plays a key role in mitochondria respiratory activity, maintains normal cognitive function, and identifies the hippocampus as a potential therapeutic target for BTHS.


Asunto(s)
Cardiolipinas/metabolismo , Disfunción Cognitiva/genética , Hipocampo/metabolismo , Factores de Transcripción/genética , Aciltransferasas , Animales , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Lisofosfolípidos , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sinaptofisina/metabolismo , Factores de Transcripción/metabolismo
7.
Neurochem Int ; 108: 266-271, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28461173

RESUMEN

Alzheimer's disease pathology includes, beside neuronal damage, reactive gliosis and reduced blood-brain barrier (BBB) integrity. Microglia are intimately associated with the BBB and upon AD pathology, pro-inflammatory responses of microglia could contribute to BBB damage. To study whether microglia can directly affect BBB integrity, the effects of amyloid beta (Aß) -stimulated primary murine microglia on co-cultured mouse brain endothelial cells (bEnd3) and murine astrocyte cultures were assessed. We also assessed whether microglial phenotype modulation via poly(ADP-ribose) polymerase-1 (PARP-1) inhibition/ablation can reverse microglial impact on these BBB forming cells. Unstimulated microglia promoted expression of tight junction proteins (TJPs), zonula ocluden-1 (ZO-1) and occludin in co-cultured endothelia cells, whereas Aß-stimulated microglia reduced endothelial expression of ZO-1 and occludin. Astrocytes co-cultured with microglia showed elevated glial fibrillary acidic protein (GFAP) expression, which was further increased if microglia had been stimulated with Aß. Aß induced microglial release of nitric oxide (NO) and tumour necrosis factor alpha (TNFα), which resulted in reduced endothelial expression of TJPs and increased paracellular permeability. Microglial PARP-1 inhibition attenuated these Aß-induced events. These findings demonstrate that PARP-1 mediated microglial responses (NO and TNFα) can directly reduce BBB integrity by promoting TJP degradation, increasing endothelial cell permeability and inducing astrogliosis. PARP-1 as a modulator of microglial phenotype can prevent microglial BBB damaging events, and thus is a potential therapeutic target.


Asunto(s)
Endotelio Vascular/metabolismo , Microglía/fisiología , Poli(ADP-Ribosa) Polimerasa-1/fisiología , Uniones Estrechas/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Técnicas de Cocultivo , Endotelio Vascular/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos
8.
J Neuroinflammation ; 14(1): 80, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28388927

RESUMEN

BACKGROUND: Birth cohort studies link gestational diabetes mellitus (GDM) with impaired cognitive performance in the offspring. However, the mechanisms involved are unknown. We tested the hypothesis that obesity-associated GDM induces chronic neuroinflammation and disturbs the development of neuronal circuitry resulting in impaired cognitive abilities in the offspring. METHODS: In rats, GDM was induced by feeding dams a diet high in sucrose and fatty acids. Brains of neonatal (E20) and young adult (15-week-old) offspring of GDM and lean dams were analyzed by immunohistochemistry, cytokine assay, and western blotting. Young adult offspring of GDM and lean dams went also through cognitive assessment. Cultured microglial responses to elevated glucose and/or fatty acids levels were analyzed. RESULTS: In rats, impaired recognition memory was observed in the offspring of GDM dams. GDM exposure combined with a postnatal high-fat and sucrose diet resulted in atypical inattentive behavior in the offspring. These cognitive changes correlated with reduced density and derangement of Cornu Ammonis 1 pyramidal neuronal layer, decreased hippocampal synaptic integrity, increased neuroinflammatory status, and reduced expression of CX3CR1, the microglial fractalkine receptor regulating microglial pro-inflammatory responses and synaptic pruning. Primary microglial cultures that were exposed to high concentrations of glucose and/or palmitate were transformed into an activated, amoeboid morphology with increased nitric oxide and superoxide production, and altered their cytokine release profile. CONCLUSIONS: These findings demonstrate that GDM stimulates microglial activation and chronic inflammatory responses in the brain of the offspring that persist into young adulthood. Reactive gliosis correlates positively with hippocampal synaptic decline and cognitive impairments. The elevated pro-inflammatory cytokine expression at the critical period of hippocampal synaptic maturation suggests that neuroinflammation might drive the synaptic and cognitive decline in the offspring of GDM dams. The importance of microglia in this process is supported by the reduced Cx3CR1 expression as an indication of the loss of microglial control of inflammatory responses and phagocytosis and synaptic pruning in GDM offspring.


Asunto(s)
Cognición/fisiología , Diabetes Gestacional/metabolismo , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Neuronas/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Células Cultivadas , Diabetes Gestacional/patología , Dieta Alta en Grasa/efectos adversos , Sacarosa en la Dieta/administración & dosificación , Sacarosa en la Dieta/efectos adversos , Femenino , Hipocampo/patología , Inflamación/metabolismo , Inflamación/patología , Masculino , Neuronas/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
9.
Artículo en Inglés | MEDLINE | ID: mdl-28405428

RESUMEN

BACKGROUND: Tail biting is a common and serious welfare problem in pig production, causing large economical losses. Tail docking is performed routinely in most EU countries to reduce the tail biting risk. However, tail docking is painful, and does not prevent tail biting totally. The risk factors behind tail docking are multifactorial and most analyses are based on studies using biological or epidemiological approaches. There is very little information available on how producers deal with tail biting on-farm. There are also no studies on the attitude of producers towards tail docking and tail biting in systems with long-tailed pigs. We aimed to study how farmers rate the efficiency of different measures for preventing and intervening with tail biting, when tail docking is not allowed. Furthermore, we investigated the attitudes of Finnish farmers to tail docking and tail biting. RESULTS: Respondents scored feeding-related issues to be most important for prevention of tail biting, identifying and removing the biting pig as most important intervention measures, and straw as the most important manipulable material when preventing tail biting. Tail biting was not perceived as a very serious problem by over 70 % of the respondents, even though docking is not allowed, and was reported to occur close to a level which was also considered acceptable by the respondents. Most respondents did not think it is probable they would raise tail docked pigs if it were possible, but about 21 % probably would. CONCLUSIONS: In comparison with other authors' findings, the ranking of importance of risk factors for tail biting differs between scientists and farmers, and between farmers in different cultures of pig production. In addition, the attitude towards tail biting and tail docking appears to be very different in producers with different experiences of tail docking. These results indicate that a scientist-farmer dialogue, as well as international communication is important when trying to reduce the risk of tail biting, and subsequently the need for tail docking.

10.
J Neuroinflammation ; 12: 229, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26637332

RESUMEN

BACKGROUND: The nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) is required for pro-inflammatory effects of TNFα. Our previous studies demonstrated that PARP-1 mediates TNFα-induced NF-κB activation in glia. Here, we evaluated the mechanisms by which TNFα activates PARP-1 and PARP-1 mediates NF-κB activation. METHODS: Primary cultures of mouse cortical astrocytes and microglia were treated with TNFα and suitable signaling pathway modulators (pharmacological and molecular). Outcome measures included calcium imaging, PARP-1 activation status, NF-κB transcriptional activity, DNA damage assesment and cytokine relesease profiling. RESULTS: TNFα induces PARP-1 activation in the absence of detectable DNA strand breaks, as measured by the PANT assay. TNFα-induced transcriptional activation of NF-κB requires PARP-1 enzymatic activity. Enzymatic activation of PARP-1 by TNFα was blocked in Ca(2+)-free medium, by Ca(2+) chelation with BAPTA-AM, and by D609, an inhibitor of phoshatidyl choline-specific phospholipase C (PC-PLC), but not by thapsigargin or by U73112, an inhibitor of phosphatidyl inisitol-specific PLC (PI -PLC). A TNFR1 blocking antibody reduced Ca(2+) influx and PARP-1 activation. TNFα-induced PARP-1 activation was also blocked by siRNA downregulation of ERK2 and by PD98059, an inhibitor of the MEK / ERK protein kinase cascade. Moreover, TNFα-induced NF-κB (p65) transcriptional activation was absent in cells expressing PARP-1 that lacked ERK2 phosphorylation sites, while basal NF-κB transcriptional activation increased in cells expressing PARP-1 with a phosphomimetic substitution at an ERK2 phophorylation site. CONCLUSIONS: These results suggest that TNFα induces PARP-1 activation through a signaling pathway involving TNFR1, Ca(2+) influx, activation of PC-PLC, and activation of the MEK1 / ERK2 protein kinase cascade. TNFα-induced PARP-1 activation is not associated with DNA damage, but ERK2 mediated phosphorylation of PARP-1.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/fisiología , FN-kappa B/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/fisiología , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Fosfolipasas de Tipo C/fisiología , Animales , Señalización del Calcio/efectos de los fármacos , Quelantes/farmacología , Daño del ADN , Activación Enzimática/efectos de los fármacos , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , ARN Interferente Pequeño/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Fosfolipasas de Tipo C/antagonistas & inhibidores
11.
J Neuroinflammation ; 12: 104, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-26017142

RESUMEN

BACKGROUND: To evaluate the role of NADPH oxidase-mediated reactive oxygen species (ROS) production in multiple sclerosis pathogenesis, we examined the effects of apocynin, an NADPH oxidase assembly inhibitor, on experimental autoimmune encephalomyelitis (EAE). METHODS: EAE was induced by immunization with myelin oligodendrocyte glycoprotein (MOG (35-55)) in C57BL/6 female mice. Three weeks after initial immunization, the mice were analyzed for demyelination, immune cell infiltration, and ROS production. Apocynin (30 mg/kg) was given orally once daily for the entire experimental course or after the typical onset of clinical symptom (15 days after first MOG injection). RESULTS: Clinical signs of EAE first appeared on day 11 and reached a peak level on day 19 after the initial immunization. The daily clinical symptoms of EAE mice were profoundly reduced by apocynin. The apocynin-mediated inhibition of the clinical course of EAE was accompanied by suppression of demyelination, reduced infiltration by encephalitogenic immune cells including CD4, CD8, CD20, and F4/80-positive cells. Apocynin reduced MOG-induced pro-inflammatory cytokines in cultured microglia. Apocynin also remarkably inhibited EAE-associated ROS production and blood-brain barrier (BBB) disruption. Furthermore, the present study found that post-treatment with apocynin also reduced the clinical course of EAE and spinal cord demyelination. CONCLUSIONS: These results demonstrate that apocynin inhibits the clinical features and neuropathological changes associated with EAE. Therefore, the present study suggests that inhibition of NADPH oxidase activation by apocynin may have a high therapeutic potential for treatment of multiple sclerosis pathogenesis.


Asunto(s)
Encéfalo/enzimología , Encefalomielitis Autoinmune Experimental/complicaciones , Leucoencefalopatías/etiología , Leucoencefalopatías/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidad , NADPH Oxidasas/metabolismo , Médula Espinal/enzimología , Acetofenonas/farmacología , Acetofenonas/uso terapéutico , Animales , Animales Recién Nacidos , Barrera Hematotesticular/fisiopatología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Leucoencefalopatías/tratamiento farmacológico , Leucoencefalopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , NADPH Oxidasas/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Índice de Severidad de la Enfermedad
12.
J Neurosci ; 35(8): 3384-96, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716838

RESUMEN

Clearing cellular debris after brain injury represents an important mechanism in regaining tissue homeostasis and promoting functional recovery. Triggering receptor expressed on myeloid cells-2 (TREM2) is a newly identified receptor expressed on microglia and is thought to phagocytose damaged brain cells. The precise role of TREM2 during ischemic stroke has not been fully understood. We explore TREM2 in both in vitro and in vivo stroke models and identify a potential endogenous TREM2 ligand. TREM2 knockdown in microglia reduced microglial activation to an amoeboid phenotype and decreased the phagocytosis of injured neurons. Phagocytosis and infarcted brain tissue resorption was reduced in TREM2 knock-out (KO) mice compared with wild-type (WT) mice. TREM2 KO mice also had worsened neurological recovery and decreased viable brain tissue in the ipsilateral hemisphere. The numbers of activated microglia and phagocytes in TREM2 KO mice were decreased compared with WT mice, and foamy macrophages were nearly absent in the TREM2 KO mice. Postischemia, TREM2 was highly expressed on microglia and TREM2-Fc fusion protein (used as a probe to identify potential TREM2 binding partners) bound to an unknown TREM2 ligand that colocalized to neurons. Oxygen glucose deprivation-exposed neuronal media, or cellular fractions containing nuclei or purified DNA, but not cytosolic fractions, stimulated signaling through TREM2. TREM2-Fc fusion protein pulled down nucleic acids from ischemic brain lysate. These findings establish the relevance of TREM2 in the phagocytosis of the infarcted brain and emphasize its role in influencing neurological outcomes following stroke. Further, nucleic acids may be one potential ligand of TREM2 in brain ischemia.


Asunto(s)
Infarto de la Arteria Cerebral Media/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Fagocitosis , Receptores Inmunológicos/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Células Espumosas/metabolismo , Células Espumosas/patología , Infarto de la Arteria Cerebral Media/patología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Neuronas/metabolismo , Neuronas/patología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética
13.
Mol Cell Neurosci ; 64: 95-103, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25553923

RESUMEN

Transcription factors are known to play multiple roles in cellular function. Investigators report that factors such as early growth response (Egr) protein and nuclear factor kappa B (NF-κB) are activated in the brain during cancer, brain injury, inflammation, and/or memory. To explore NF-κB activity further, we investigated the transcriptomes of hippocampal slices following electrical stimulation of NF-κB p50 subunit knockout mice (p50-/-) versus their controls (p50+/+). We found that the early growth response gene Egr-2 was upregulated by NF-κB activation, but only in p50+/+ hippocampal slices. We then stimulated HeLa cells and primary cortical neurons with tumor necrosis factor alpha (TNFα) to activate NF-κB and increase the expression of Egr-2. The Egr-2 promoter sequence was analyzed for NF-κB binding sites and chromatin immunoprecipitation (ChIP) assays were performed to confirm promoter occupancy in vivo. We discovered that NF-κB specifically binds to an NF-κB consensus binding site within the proximal promoter region of Egr-2. Luciferase assay demonstrated that p50 was able to transactivate the Egr-2 promoter in vitro. Small interfering RNA (siRNA)-mediated p50 knockdown corroborated other Egr-2 expression studies. We show for the first time a novel link between NF-κB activation and Egr-2 expression with Egr-2 expression directly controlled by the transcriptional activity of NF-κB.


Asunto(s)
Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Activación Transcripcional , Animales , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Células HeLa , Hipocampo/metabolismo , Hipocampo/fisiología , Humanos , Ratones , Subunidad p50 de NF-kappa B/genética , Regiones Promotoras Genéticas , Unión Proteica
14.
J Neuroinflammation ; 10: 49, 2013 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-23607899

RESUMEN

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis characterized by entry of activated T cells and antigen presenting cells into the central nervous system and subsequent autoimmune destruction of nerve myelin. Previous studies revealed that non-selective inhibition of poly(ADP-ribose) polymerases (PARPs) 1 and 2 protect against neuroinflammation and motor dysfunction associated with EAE, but the role of the PARP-2 isoform has not yet been investigated selectively. RESULTS: EAE was induced in mice lacking PARP-2, and neurological EAE signs, blood-spine barrier (BSB) permeability, demyelination and inflammatory infiltration were monitored for 35 days after immunization. Mice lacking PARP-2 exhibited significantly reduced overall disease burden and peak neurological dysfunction. PARP-2 deletion also significantly delayed EAE onset and reduced BSB permeability, demyelination and central nervous system (CNS) markers of proinflammatory Th1 and Th17 T helper lymphocytes. CONCLUSIONS: This study represents the first description of a significant role for PARP-2 in neuroinflammation and neurological dysfunction in EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Inflamación/patología , Enfermedades del Sistema Nervioso/patología , Poli(ADP-Ribosa) Polimerasas/fisiología , Animales , Barrera Hematonerviosa/fisiología , Enfermedades Desmielinizantes/patología , Encefalomielitis Autoinmune Experimental/complicaciones , Técnica del Anticuerpo Fluorescente , Inflamación/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades del Sistema Nervioso/etiología , Infiltración Neutrófila/fisiología , Poli(ADP-Ribosa) Polimerasas/genética , Linfocitos T Colaboradores-Inductores/fisiología , Células TH1/fisiología
15.
Biochim Biophys Acta ; 1833(8): 1985-91, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23597856

RESUMEN

NF-κB is a transcription factor that integrates pro-inflammatory and pro-survival responses in diverse cell types. The activity of NF-κB is regulated in part by acetylation of its p65 subunit at lysine 310, which is required for transcription complex formation. De-acetylation at this site is performed by sirtuin 1(SIRT1) and possibly other sirtuins in an NAD(+) dependent manner, such that SIRT1 inhibition promotes NF-κB transcriptional activity. It is unknown, however, whether changes in NAD(+) levels can influence p65 acetylation and cellular inflammatory responses. Poly(ADP-ribose)-1 (PARP-1) is an abundant nuclear enzyme that consumes NAD(+) in the process of forming (ADP-ribose)polymers on target proteins, and extensive PARP-1 activation can reduce intracellular NAD(+) concentrations. Here we tested the idea that PARP-1 activation can regulate NF-κB transcriptional activity by reducing NAD(+) concentrations and thereby inhibiting de-acetylation of p65. Primary astrocyte cultures were treated with the alkylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) to induce PARP-1 activation. This resulted in sustained acetylation of p65 and increased NF-κB transcriptional activity as monitored by a κB-driven eGFP reporter gene. These effects of MNNG were negated by a PARP-1 inhibitor, in PARP-1(-/-) cells, and in PARP-1(-/-) cells transfected with a catalytically inactive PARP-1 construct, thus confirming that these effects are mediated by PARP-1 catalytic activity. The effects of PARP-1 activation were replicated by a SIRT1 inhibitor, EX-527, and were reversed by exogenous NAD(+). These findings demonstrate that PARP-1-induced changes in NAD(+) levels can modulate NF-κB transcriptional activity through effects on p65 acetylation.


Asunto(s)
NAD/deficiencia , FN-kappa B/genética , FN-kappa B/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , eIF-2 Quinasa/metabolismo , Acetilación/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Astrocitos/metabolismo , Astrocitos/fisiología , Carbazoles/farmacología , Células Cultivadas , Humanos , Metilnitronitrosoguanidina/farmacología , Ratones , NAD/genética , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Sirtuina 1/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Transfección , eIF-2 Quinasa/genética
16.
J Neuroinflammation ; 9: 225, 2012 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-22998689

RESUMEN

Diabetic patients who attempt strict management of blood glucose levels frequently experience hypoglycemia. Severe and prolonged hypoglycemia causes neuronal death and cognitive impairment. There is no effective tool for prevention of these unwanted clinical sequelae. Minocycline, a second-generation tetracycline derivative, has been recognized as an anti-inflammatory and neuroprotective agent in several animal models such as stroke and traumatic brain injury. In the present study, we tested whether minocycline also has protective effects on hypoglycemia-induced neuronal death and cognitive impairment. To test our hypothesis we used an animal model of insulin-induced acute hypoglycemia. Minocycline was injected intraperitoneally at 6 hours after hypoglycemia/glucose reperfusion and injected once per day for the following 1 week. Histological evaluation for neuronal death and microglial activation was performed from 1 day to 1 week after hypoglycemia. Cognitive evaluation was conducted 6 weeks after hypoglycemia. Microglial activation began to be evident in the hippocampal area at 1 day after hypoglycemia and persisted for 1 week. Minocycline injection significantly reduced hypoglycemia-induced microglial activation and myeloperoxidase (MPO) immunoreactivity. Neuronal death was significantly reduced by minocycline treatment when evaluated at 1 week after hypoglycemia. Hypoglycemia-induced cognitive impairment is also significantly prevented by the same minocycline regimen when subjects were evaluated at 6 weeks after hypoglycemia. Therefore, these results suggest that delayed treatment (6 hours post-insult) with minocycline protects against microglial activation, neuronal death and cognitive impairment caused by severe hypoglycemia. The present study suggests that minocycline has therapeutic potential to prevent hypoglycemia-induced brain injury in diabetic patients.


Asunto(s)
Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Hipoglucemia/complicaciones , Minociclina/uso terapéutico , Neuronas/efectos de los fármacos , Análisis de Varianza , Animales , Glucemia/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Antígeno CD11b/metabolismo , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Fluoresceínas , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipoglucemia/inducido químicamente , Hipoglucemia/tratamiento farmacológico , Hipoglucemia/patología , Hipoglucemiantes/toxicidad , Insulina/toxicidad , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Movimiento/efectos de los fármacos , Neuronas/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Compuestos Orgánicos , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley
17.
J Neuroinflammation ; 9: 182, 2012 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-22830525

RESUMEN

BACKGROUND: Recurrent/moderate (R/M) hypoglycemia is common in type 1 diabetes. Although mild or moderate hypoglycemia is not life-threatening, if recurrent, it may cause cognitive impairment. In the present study, we sought to determine whether R/M hypoglycemia leads to neuronal death, dendritic injury, or cognitive impairment. METHODS: The experiments were conducted in normal and in diabetic rats. Rats were subjected to moderate hypoglycemia by insulin without anesthesia. Oxidative stress was evaluated by 4-Hydroxy-2-nonenal immunostaining and neuronal death was determined by Fluoro-Jade B staining 7 days after R/M hypoglycemia. To test whether oxidative injury caused by NADPH oxidase activation, an NADPH oxidase inhibitor, apocynin, was used. Cognitive function was assessed by Barnes maze and open field tests at 6 weeks after R/M hypoglycemia. RESULTS: The present study found that oxidative injury was detected in the dendritic area of the hippocampus after R/M hypoglycemia. Sparse neuronal death was found in the cortex, but no neuronal death was detected in the hippocampus. Significant cognitive impairment and thinning of the CA1 dendritic region was detected 6 weeks after hypoglycemia. Oxidative injury, cognitive impairment, and hippocampal thinning after R/M hypoglycemia were more severe in diabetic rats than in non-diabetic rats. Oxidative damage in the hippocampal CA1 dendritic area and microglial activation were reduced by the NADPH oxidase inhibitor, apocynin. CONCLUSION: The present study suggests that oxidative injury of the hippocampal CA1 dendritic region by R/M hypoglycemia is associated with chronic cognitive impairment in diabetic patients. The present study further suggests that NADPH oxidase inhibition may prevent R/M hypoglycemia-induced hippocampal dendritic injury.


Asunto(s)
Trastornos del Conocimiento/etiología , Dendritas/patología , Hipocampo/patología , Hipoglucemia/complicaciones , Microglía/patología , Animales , Glucemia/metabolismo , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Dendritas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Hipocampo/metabolismo , Hipoglucemia/metabolismo , Hipoglucemia/patología , Masculino , Microglía/metabolismo , Ratas , Ratas Sprague-Dawley , Recurrencia
18.
J Neuroinflammation ; 9: 31, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22335939

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) induces activation of microglia. Activated microglia can in turn increase secondary injury and impair recovery. This innate immune response requires hours to days to become fully manifest, thus providing a clinically relevant window of opportunity for therapeutic intervention. Microglial activation is regulated in part by poly(ADP-ribose) polymerase-1 (PARP-1). Inhibition of PARP-1 activity suppresses NF-kB-dependent gene transcription and thereby blocks several aspects of microglial activation. Here we evaluated the efficacy of a PARP inhibitor, INO-1001, in suppressing microglial activation after cortical impact in the rat. METHODS: Rats were subjected to controlled cortical impact and subsequently treated with 10 mg/kg of INO-1001 (or vehicle alone) beginning 20 - 24 hours after the TBI. Brains were harvested at several time points for histological evaluation of inflammation and neuronal survival, using markers for microglial activation (morphology and CD11b expression), astrocyte activation (GFAP), and neuronal survival (NeuN). Rats were also evaluated at 8 weeks after TBI using measures of forelimb dexterity: the sticky tape test, cylinder test, and vermicelli test. RESULTS: Peak microglial and astrocyte activation was observed 5 to 7 days after this injury. INO-1001 significantly reduced microglial activation in the peri-lesion cortex and ipsilateral hippocampus. No rebound inflammation was observed in rats that were treated with INO-1001 or vehicle for 12 days followed by 4 days without drug. The reduced inflammation was associated with increased neuronal survival in the peri-lesion cortex and improved performance on tests of forelimb dexterity conducted 8 weeks after TBI. CONCLUSIONS: Treatment with a PARP inhibitor for 12 days after TBI, with the first dose given as long as 20 hours after injury, can reduce inflammation and improve histological and functional outcomes.


Asunto(s)
Lesiones Encefálicas/patología , Indoles/uso terapéutico , Microglía/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo , Análisis de Varianza , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Antígeno CD11b/metabolismo , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Miembro Anterior/fisiopatología , Lateralidad Funcional , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Inyecciones Intraperitoneales/métodos , Masculino , Microglía/fisiología , Destreza Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
19.
J Neuroinflammation ; 9: 22, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22272874

RESUMEN

BACKGROUND: Microglia, the resident immune cells of the brain, have been implicated in brain injury and various neurological disorders. However, their precise roles in different pathophysiological situations remain enigmatic and may range from detrimental to protective. Targeting the delivery of biologically active compounds to microglia could help elucidate these roles and facilitate the therapeutic modulation of microglial functions in neurological diseases. METHODS: Here we employ primary cell cultures and stereotaxic injections into mouse brain to investigate the cell type specific localization of semiconductor quantum dots (QDs) in vitro and in vivo. Two potential receptors for QDs are identified using pharmacological inhibitors and neutralizing antibodies. RESULTS: In mixed primary cortical cultures, QDs were selectively taken up by microglia; this uptake was decreased by inhibitors of clathrin-dependent endocytosis, implicating the endosomal pathway as the major route of entry for QDs into microglia. Furthermore, inhibiting mannose receptors and macrophage scavenger receptors blocked the uptake of QDs by microglia, indicating that QD uptake occurs through microglia-specific receptor endocytosis. When injected into the brain, QDs were taken up primarily by microglia and with high efficiency. In primary cortical cultures, QDs conjugated to the toxin saporin depleted microglia in mixed primary cortical cultures, protecting neurons in these cultures against amyloid beta-induced neurotoxicity. CONCLUSIONS: These findings demonstrate that QDs can be used to specifically label and modulate microglia in primary cortical cultures and in brain and may allow for the selective delivery of therapeutic agents to these cells.


Asunto(s)
Encéfalo/citología , Microglía/fisiología , Puntos Cuánticos , Péptidos beta-Amiloides/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Receptor 1 de Quimiocinas CX3C , Proteínas de Unión al Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Corteza Cerebral/citología , Clatrina/metabolismo , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Inmunotoxinas/farmacología , Mananos/farmacología , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fragmentos de Péptidos/farmacología , Poli I/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Quimiocina/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Saporinas , Técnicas Estereotáxicas , Factores de Tiempo
20.
J Neuroinflammation ; 8: 152, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-22051244

RESUMEN

BACKGROUND: Amyloid ß (Aß) accumulates in Alzheimer's disease (AD) brain. Microglial activation also occurs in AD, and this inflammatory response may contribute to disease progression. Microglial activation can be induced by Aß, but the mechanisms by which this occurs have not been defined. The nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) regulates microglial activation in response to several stimuli through its interactions with the transcription factor, NF-κB. The purpose of this study was to evaluate whether PARP-1 activation is involved in Aß-induced microglial activation, and whether PARP-1 inhibition can modify microglial responses to Aß. METHODS: hAPP(J20) mice, which accumulate Aß with ageing, were crossed with PARP-1(-/-) mice to assess the effects of PARP-1 depletion on microglial activation, hippocampal synaptic integrity, and cognitive function. Aß peptide was also injected into brain of wt and PARP-1(-/-) mice to directly determine the effects of PARP-1 on Aß-induced microglial activation. The effect of PARP-1 on Aß-induced microglial cytokine production and neurotoxicity was evaluated in primary microglia cultures and in microglia-neuron co-cultures, utilizing PARP-1(-/-) cells and a PARP-1 inhibitor. NF-κB activation was evaluated in microglia infected with a lentivirus reporter gene. RESULTS: The hAPP(J20) mice developed microglial activation, reduced hippocampal CA1 calbindin expression, and impaired novel object recognition by age 6 months. All of these features were attenuated in hAPP(J20)/PARP-1(-/-) mice. Similarly, Aß(1-42) injected into mouse brain produced a robust microglial response in wild-type mice, and this was blocked in mice lacking PARP-1 expression or activity. Studies using microglial cultures showed that PARP-1 activity was required for Aß-induced NF-κB activation, morphological transformation, NO release, TNFα release, and neurotoxicity. Conversely, PARP-1 inhibition increased release of the neurotrophic factors TGFß and VEGF, and did not impair microglial phagocytosis of Aß peptide. CONCLUSIONS: These results identify PARP-1 as a requisite and previously unrecognized factor in Aß-induced microglial activation, and suggest that the effects of PARP-1 are mediated, at least in part, by its interactions with NF-κB. The suppression of Aß-induced microglial activation and neurotoxicity by PARP-1 inhibition suggests this approach could be useful in AD and other disorders in which microglial neurotoxicity may contribute.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Microglía/efectos de los fármacos , Microglía/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Calbindinas , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , FN-kappa B/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fagocitosis/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...