Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Curr Med Chem ; 2023 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-36815654

RESUMO

Epilepsy is a chronic neurological degenerative disease with a high incidence, affecting all age groups. Refractory Epilepsy (RE) occurs in approximately 30-40% of cases with a higher risk of sudden unexpected death in epilepsy (SUDEP). Recent studies have shown that spontaneous seizures developed in epilepsy can be related to an increase in oxidative stress and reactive oxygen derivatives (ROS) production. Increasing ROS concentration causes lipid peroxidation, protein oxidation, destruction of nuclear genetic material, enzyme inhibition, and cell death by a mechanism known as "ferroptosis" (Fts). Inactivation of glutathione peroxidase 4 (GPX4) induces Fts, while oxidative stress is linked with increased intracellular free iron (Fe+2) concentration. Fts is also a non-apoptotic programmed cell death mechanism, where a hypoxia-inducible factor 1 alpha (HIF-141) dependent hypoxic stress-like condition appears to occur with accumulation of iron and cytotoxic ROS in affected cells. Assuming convulsive crises as hypoxic stress, repetitive convulsive/hypoxic stress can be an effective inducer of the "epileptic heart" (EH), which is characterized by altered autonomic function and a high risk of malignant or fatal bradycardia. We previously reported that experimental recurrent seizures induce cardiomyocyte Fts associated with SUDEP. Furthermore, several genes related to Fts and hypoxia have recently been identified in acute myocardial infarction. An emerging theme from recent studies indicates that inhibition of GPX4 through modulating expression or activities of the xCT antiporter system (SLC7A11) governs cell sensitivity to oxidative stress from ferroptosis. Furthermore, during hypoxia, an increased expression of stress transcriptional factor ATF3 can promote Fts induced by erastin in a HIF-141-dependent manner. We propose that inhibition of Fts with ROS scavengers, iron chelators, antioxidants, and transaminase inhibitors could provide a therapeutic effect in epilepsy and improve the prognosis of SUDEP risk by protecting the heart from ferroptosis.

2.
Curr Med Chem ; 30(35): 3993-4031, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36200215

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by marked cognitive decline, memory loss, and spatio-temporal troubles and, in severe cases, lack of recognition of family members. Neurological symptoms, cognitive disturbances, and the inflammatory frame due to COVID-19, together with long-term effects, have fueled renewed interest in AD based on similar damage. COVID-19 also caused the acceleration of AD symptom onset. In this regard, the morbidity and mortality of COVID-19 were reported to be increased in patients with AD due to multiple pathological changes such as excessive expression of the viral receptor angiotensin-converting enzyme 2 (ACE2), comorbidities such as diabetes, hypertension, or drug-drug interactions in patients receiving polypharmacy and the high presence of proinflammatory molecules. Furthermore, the release of cytokines, neuroinflammation, oxidative stress, and ferroptosis in both diseases showed common underlying mechanisms, which together worsen the clinical picture and prognosis of these patients.


Assuntos
Doença de Alzheimer , COVID-19 , Ferroptose , Doenças Neurodegenerativas , Humanos , Doença de Alzheimer/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neuroinflamatórias , Estresse Oxidativo
3.
Acta bioquím. clín. latinoam ; 56(4): 490-513, dic. 2022. graf
Artigo em Espanhol | LILACS-Express | LILACS, BINACIS | ID: biblio-1439101

RESUMO

Resumen El hierro (Fe) es un elemento vital para casi todos los organismos debido a su facilidad para donar y aceptar electrones. Es cofactor de muchas proteínas y enzimas necesarias para la adecuada utilización del oxígeno y la generación de energía. Su desregulación se relaciona a procesos de estrés oxidativo y muerte celular mediada por Fe(II) denominada ferroptosis. Las células de mamíferos utilizan múltiples mecanismos para garantizar la adquisición del hierro como nutriente esencial, que se encuentra oxidado [Fe(III)], y que debe ser reducido a Fe(II) para su adecuada utilización intracelular. Cada etapa de transferencia del hierro a través de las membranas biológicas exige una reconversión de su estado de oxidado a reducido y viceversa, dependiendo del paso metabólico implicado. La distorsión de dichos procesos se asocia con varias enfermedades: desde la deficiencia de hierro debida a defectos en la adquisición o distribución del metal, que causa anemia, a la sobrecarga de hierro que resulta de una absorción excesiva de hierro o en una utilización defectuosa, que causa una sobreoferta de Fe(II) en los tejidos y que lleva a un daño oxidativo y a la muerte celular. Existen múltiples mecanismos regulatorios que en conjunto aseguran el equilibrio en la homeostasis del hierro. Esta actualización describe los avances recientes en las vías reguladoras del hierro, así como en los mecanismos subyacentes al tráfico de dicho elemento desde su absorción, principalmente biodistribución y su uso intracelular, quizás el área más importante donde se define su adecuada utilización o la muerte celular por ferroptosis.


Abstract Iron (Fe) is a vital element for almost all organisms due to its ability to donate and accept electrons with relative ease. It serves as a cofactor for many proteins and enzymes necessary for the proper use of oxygen and energy generation, and its deregulation is related to the processes of oxidative stress and iron-mediated cell death called ferroptosis. Mammalian cells use multiple mechanisms to ensure the acquisition of iron as an essential nutrient, which is normally oxidised in the form of Fe(III) and must be reduced to Fe(II) for adequate intracellular use. Each stage of iron transfer across biological membranes requires a reconversion of its state from oxidised to reduced and vice versa, depending on the metabolic step involved. Distortion of these processes is associated with various diseases, such as iron deficiency due to defects in the acquisition or distribution of the metal that causes anemia, as well as iron overload from excessive iron absorption or defective use, which results in an oversupply of Fe(II) in tissues leading to oxidative damage and cell death. There are multiple regulatory mechanisms that together ensure the balance in iron homeostasis. This update describes the recent advances in the iron regulatory pathways, as well as in the mechanisms underlying iron trafficking from its absorption, mainly biodistribution and its intracellular use, perhaps the most important area where its adequate utilisation or cell death by ferroptosis is defined.


Resumo O ferro (Fe) é um elemento vital para quase todos os organismos devido à sua capacidade de doar e aceitar elétrons com relativa facilidade. O ferro serve como cofator para muitas proteínas e enzimas necessárias para o uso adequado do oxigênio e geração de energia, e a sua desregulação está relacionada a processos de estresse oxidativo e morte celular mediada por Fe(II) denominado ferroptose. As células de mamíferos utilizam múltiplos mecanismos para garantir a aquisição de ferro como nutriente essencial, que normalmente é oxidado na forma de Fe(III) e deve ser reduzido a Fe(II) para o uso intracelular adequado. Cada estágio de transferência de Fe através das membranas biológicas requer uma reconversão de seu estado de oxidado para reduzido e vice-versa, dependendo da etapa metabólica envolvida. A distorção desses processos está associada a várias doenças: desde a deficiência de ferro devido a defeitos na aquisição ou distribuição do metal que causa a anemia, até a sobrecarga de ferro resultante da absorção excessiva de ferro ou utilização defeituosa, que causa um excesso de oferta de Fe(II) nos tecidos levando ao dano oxidativo e morte celular. Existem múltiplos mecanismos regulatórios que juntos garantem o equilíbrio na homeostase do ferro. Esta atualização descreve os avanços recentes nas vias reguladoras do ferro, bem como nos mecanismos subjacentes ao tráfico deste elemento desde a sua absorção, principalmente biodistribuição e seu uso intracelular, talvez a área mais importante onde sua utilização adequada ou morte celular por ferroptose é definido.

4.
Epilepsia Open ; 7 Suppl 1: S34-S46, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-34542938

RESUMO

The multidrug-resistance (MDR) phenotype is typically observed in patients with refractory epilepsy (RE) whose seizures are not controlled despite receiving several combinations of more than two antiseizure medications (ASMs) directed against different ion channels or neurotransmitter receptors. Since the use of bromide in 1860, more than 20 ASMs have been developed; however, historically ~30% of cases of RE with MDR phenotype remains unchanged. Irrespective of metabolic biotransformation, the biodistribution of ASMs and their metabolites depends on the functional expression of some ATP-binding cassette transporters (ABC-t) in different organs, such as the blood-brain barrier (BBB), bowel, liver, and kidney, among others. ABC-t, such as P-glycoprotein (P-gp), multidrug resistance-associated protein (MRP-1), and breast cancer-resistance protein (BCRP), are mainly expressed in excretory organs and play a critical role in the pharmacokinetics (PK) of all drugs. The transporter hypothesis can explain pharmacoresistance to a broad spectrum of ASMs, even when administered simultaneously. Since ABC-t expression can be induced by hypoxia, inflammation, or seizures, a high frequency of uncontrolled seizures increases the risk of RE. These stimuli can induce ABC-t expression in excretory organs and in previously non-expressing (electrically responsive) cells, such as neurons or cardiomyocytes. In this regard, an alternative mechanism to the classical pumping function of P-gp indicates that P-gp activity can also produce a significant reduction in resting membrane potential (ΔΨ0 = -60 to -10 mV). P-gp expression in neurons and cardiomyocytes can produce membrane depolarization and participate in epileptogenesis, heart failure, and sudden unexpected death in epilepsy. On this basis, ABC-t play a peripheral role in controlling the PK of ASMs and their access to the brain and act at a central level, favoring neuronal depolarization by mechanisms independent of ion channels or neurotransmitters that current ASMs cannot control.


Assuntos
Epilepsia , Proteínas de Neoplasias , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/uso terapêutico , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/uso terapêutico , Epilepsia/tratamento farmacológico , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/uso terapêutico , Convulsões/tratamento farmacológico , Distribuição Tecidual
5.
Front Neurol ; 12: 609236, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33643194

RESUMO

Uncontrolled repetitive generalized tonic-clonic seizures (GTCS) are the main risk factor for sudden unexpected death in epilepsy (SUDEP). GTCS can be observed in models such as Pentylenetetrazole kindling (PTZ-K) or pilocarpine-induced Status Epilepticus (SE-P), which share similar alterations in cardiac function, with a high risk of SUDEP. Terminal cardiac arrhythmia in SUDEP can develop as a result of a high rate of hypoxic stress-induced by convulsions with excessive sympathetic overstimulation that triggers a neurocardiogenic injury, recently defined as "Epileptic Heart" and characterized by heart rhythm disturbances, such as bradycardia and lengthening of the QT interval. Recently, an iron overload-dependent form of non-apoptotic cell death called ferroptosis was described at the brain level in both the PTZ-K and SE-P experimental models. However, seizure-related cardiac ferroptosis has not yet been reported. Iron overload cardiomyopathy (IOC) results from the accumulation of iron in the myocardium, with high production of reactive oxygen species (ROS), lipid peroxidation, and accumulation of hemosiderin as the final biomarker related to cardiomyocyte ferroptosis. Iron overload cardiomyopathy is the leading cause of death in patients with iron overload secondary to chronic blood transfusion therapy; it is also described in hereditary hemochromatosis. GTCS, through repeated hypoxic stress, can increase ROS production in the heart and cause cardiomyocyte ferroptosis. We hypothesized that iron accumulation in the "Epileptic Heart" could be associated with a terminal cardiac arrhythmia described in the IOC and the development of state-potentially in the development of SUDEP. Using the aforementioned PTZ-K and SE-P experimental models, after SUDEP-related repetitive GTCS, we observed an increase in the cardiac expression of hypoxic inducible factor 1α, indicating hypoxic-ischemic damage, and both necrotic cells and hemorrhagic areas were related to the possible hemosiderin production in the PTZ-K model. Furthermore, we demonstrated for the first time an accumulation of hemosiderin in the heart in the SE-P model. These results suggest that uncontrolled recurrent seizures, as described in refractory epilepsy, can give rise to high hypoxic stress in the heart, thus inducing hemosiderin accumulation as in IOC, and can act as an underlying hidden mechanism contributing to the development of a terminal cardiac arrhythmia in SUDEP. Because iron accumulation in tissues can be detected by non-invasive imaging methods, cardiac iron overload in refractory epilepsy patients could be treated with chelation therapy to reduce the risk of SUDEP.

6.
Epilepsy Behav ; 121(Pt B): 106590, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-31706919

RESUMO

Sudden unexpected death in epilepsy (SUDEP) is the major cause of death that affects patients with epilepsy. The risk of SUDEP increases according to the frequency and severity of uncontrolled seizures; therefore, SUDEP risk is higher in patients with refractory epilepsy (RE), in whom most antiepileptic drugs (AEDs) are ineffective for both seizure control and SUDEP prevention. Consequently, RE and SUDEP share a multidrug resistance (MDR) phenotype, which is mainly associated with brain overexpression of ABC-transporters such as P-glycoprotein (P-gp). The activity of P-gp can also contribute to membrane depolarization and affect the normal function of neurons and cardiomyocytes. Other molecular regulators of membrane potential are the inwardly rectifying potassium channels (Kir), whose genetic variants have been related to both epilepsy and heart dysfunctions. Although it has been suggested that dysfunctions of the cardiac, respiratory, and brainstem arousal systems are the causes of SUDEP, the molecular basis for explaining its dysfunctions remain unknown. In rats, repetitive seizures or status epilepticus induced high expression of P-gp and loss Kir expression in the brain and heart, and promoted membrane depolarization, malignant bradycardia, and the high rate of mortality. Here we reviewed clinical and experimental evidences suggesting that abnormal expression of depolarizing/repolarizing factors as P-gp and Kir could favor persistent depolarization of membranes without any rapid functional recovery capacity. This condition induced by convulsive stress could be the molecular mechanism leading to acquired severe bradycardia, as an ineffective heart response generating the appropriate scenario for SUDEP development. This article is part of the Special Issue "NEWroscience 2018".


Assuntos
Epilepsia , Morte Súbita Inesperada na Epilepsia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Morte Súbita/etiologia , Epilepsia/complicações , Humanos , Potássio , Ratos , Fatores de Risco
7.
Biochim Biophys Acta Gen Subj ; 1864(4): 129522, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31945406

RESUMO

BACKGROUND: Osteosarcoma (OS) is the most frequent malignant bone tumor, affecting predominantly children and young adults. Metastases are a major clinical challenge in OS. In this context, 20% of OS patients are diagnosed with metastatic OS, but near 80% of all OS patients could present non-detectable micrometastases at the moment of diagnosis. METHODS: Osteogenic differentiation; doxorubicin exclusion assay; fluorescence microscopy; RT-qPCR; proteomic analysis. RESULTS: Our results suggest that metastatic OS cells possess a diminished osteoblastic differentiation potential with a gain of metastatic traits like the capacity to modify intracellular localization of chemodrugs and higher levels of expression of stemness-related genes. On the opposite hand, non-metastatic OS cells possess bone-associated traits like higher osteoblastic differentiation and also an osteoblastic-inducer secretome. OS cells also differ in the nature of their interaction with mesenchymal stem cells (MSCs), with opposites impacts on MSCs phenotype and behavior. CONCLUSIONS: All this suggests that a major trait acquired by metastatic cells is a switch into a stem-like state that could favor its survival in the pulmonary niche, opening new possibilities for personalized chemotherapeutic schemes. GENERAL SIGNIFICANCE: Our work provides new insights regarding differences among metastatic and non-metastatic OS cells, with particular emphasis on differentiation potential, multidrug resistance and interaction with MSCs.


Assuntos
Neoplasias Ósseas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteossarcoma/metabolismo , Antibióticos Antineoplásicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/patologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/secundário , Fenótipo , Relação Estrutura-Atividade , Células Tumorais Cultivadas
8.
Int J Mol Sci ; 20(8)2019 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-31010037

RESUMO

Mesenchymal stem cells (MSCs) represent an interesting population due to their capacity to release a variety of cytokines, chemokines, and growth factors, and due to their motile nature and homing ability. MSCs can be isolated from different sources, like adipose tissue or bone marrow, and have the capacity to differentiate, both in vivo and in vitro, into adipocytes, chondrocytes, and osteoblasts, making them even more interesting in the regenerative medicine field. Tumor associated stroma has been recognized as a key element in tumor progression, necessary for the biological success of the tumor, and MSCs represent a functionally fundamental part of this associated stroma. Exosomes represent one of the dominant signaling pathways within the tumor microenvironment. Their biology raises high interest, with implications in different biological processes involved in cancer progression, such as the formation of the pre-metastatic niche. This is critical during the metastatic cascade, given that it is the formation of a permissive context that would allow metastatic tumor cells survival within the new environment. In this context, we explored the role of exosomes, particularly MSCs-derived exosomes as direct or indirect modulators. All this points out a possible new tool useful for designing better treatment and detection strategies for metastatic progression, including the management of chemoresistance.


Assuntos
Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Metástase Neoplásica/patologia , Animais , Humanos , Tropismo , Microambiente Tumoral
9.
Blood Res ; 50(3): 154-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26457282

RESUMO

BACKGROUND: Tyrosine kinase inhibitors (TKIs) are the recommended treatment for patients with chronic myeloid leukemia (CML). The MDR1/ABCB1 gene plays a role in resistance to a wide spectrum of drugs, including TKIs. However, the association of MDR1/ABCB1 gene polymorphisms (SNPs) such as C1236T, G2677T/A, and C3435T with the clinical therapeutic evolution of CML has been poorly studied. We investigated these gene polymorphisms in CML-patients treated with imatinib, nilotinib and/or dasatinib. METHODS: ABCB1-SNPs were studied in 22 CML-patients in the chronic phase (CP) and 2 CML-patients in blast crisis (BC), all of whom were treated with TKIs, and compared with 25 healthy controls using nested-PCR and sequencing techniques. RESULTS: Seventeen different haplotypes were identified: 7 only in controls, 6 only in CML-patients, and the remaining 4 in both groups. The distribution ratios of homozygous TT-variants present on each exon between controls and CML-patients were 2.9 for exon 12, and 0.32 for the other 2 exons. Heterozygous T-variants were observed in all controls (100%) and 75% of CML-patients. Wt-haplotype (CC-GG-CC) was observed in 6 CML-patients (25%). In this wt-group, two were treated with nilotinib and reached a major molecular response. The remaining 4 cases had either a minimal or null molecular response, or developed bone marrow aplasia. CONCLUSION: Our results suggest that SNPs of the MDR1/ABCB1 gene could help to characterize the prognosis and the clinical-therapeutic evolution of CML-patients treated with TKIs. Wt-haplotype could be associated with a higher risk of developing CML, and a worse clinical-therapeutic evolution.

10.
Ann Hum Genet ; 79(6): 385-93, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26420346

RESUMO

Somatic sequence variants in the epidermal growth factor receptor (EGFR) kinase domain are associated with sensitivity to tyrosine kinase inhibitors (TKIs) in patients with nonsmall cell lung cancer (NSCLC). Patients exhibiting sequence variants in this domain that produce kinase activity enhancement, are more likely to benefit from TKIs than patients with EGFR wild-type disease. Although most NSCLC EGFR-related alleles are concentrated in a few positions, established protocols recommend sequencing EGFR exons 18-21. In this study, 21 novel somatic variants belonging to such exons in adult Argentinean patients affected with NSCLC are reported. Of these, 18 were single amino acid substitutions (SASs), occurring alone or in combination with another genetic alteration (complex cases), one was a short deletion, one was a short deletion-short insertion combination, and one was a duplication. New variants and different combinations of previously reported variants were also found. Moreover, two of the reported SASs occurred in previously unreported positions of the EGFR kinase domain. In order to characterize the new sequence variants, physicochemical, sequence and conformational analyses were also performed. A better understanding of sequence variants in NSCLC may facilitate the most appropriate treatment choice for this complex disease.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/genética , Substituição de Aminoácidos , Argentina , Éxons , Feminino , Humanos , Mutação INDEL , Masculino , Estrutura Terciária de Proteína , Deleção de Sequência
11.
Expert Rev Neurother ; 15(6): 587-96, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25961655

RESUMO

Alzheimer's disease (AD) is a degenerative neurological disorder that is the most common cause of dementia and disability in older patients. Available treatments are symptomatic in nature and are only sufficient to improve the quality of life of AD patients temporarily. A potential strategy, currently under investigation, is to target cell-signaling pathways associated with neurodegeneration, in order to decrease neuroinflammation, excitotoxicity, and to improve cognitive functions. Current review centers on the role of neuroinflammation and the specific contribution of mast cells to AD pathophysiology. The authors look at masitinib therapy and the evidence presented through preclinical and clinical trials. Dual actions of masitinib as an inhibitor of mast cell-glia axis and a Fyn kinase blocker are discussed in the context of AD pathology. Masitinib is in Phase III clinical trials for the treatment of malignant melanoma, mastocytosis, multiple myeloma, gastrointestinal cancer and pancreatic cancer. It is also in Phase II/III clinical trials for the treatment of multiple sclerosis, rheumatoid arthritis and AD. Additional research is warranted to better investigate the potential effects of masitinib in combination with other drugs employed in AD treatment.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Tiazóis/uso terapêutico , Animais , Benzamidas , Humanos , Piperidinas , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas c-kit/metabolismo , Piridinas , Tiazóis/química , Tiazóis/farmacologia
12.
Front Biosci (Elite Ed) ; 6(2): 329-40, 2014 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-24896209

RESUMO

Epilepsy is one of the most common chronic neurological disorders. Furthermore, it is associated to diminished health-related quality of life and is thus considered a major public health problem. In spite of the large number of available and ongoing development of several new antiepileptic drugs (AEDs), a high percentage of patients with epilepsy (35-40%) are resistant to pharmacotherapy. A hypothesis to explain pharmacoresistance in epilepsy suggests that overexpression of multidrug resistance proteins, such as P-glycoprotein, on the endothelium of the blood brain barrier represents a challenge for effective AED delivery and concentration levels in the brain. Proven therapeutic strategies to control pharmacoresistant epilepsy include epilepsy surgery and neuromodulation. Unfortunately, not all patients are candidates for these therapies. Nanotechnology represents an attractive strategy to overcome the limited brain access of AEDs in patients with pharmacoresistant epilepsy. This manuscript presents a review of evidences supporting this idea.


Assuntos
Portadores de Fármacos/uso terapêutico , Epilepsia/tratamento farmacológico , Epilepsia/fisiopatologia , Modelos Biológicos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Nanopartículas/uso terapêutico , Epilepsia/metabolismo , Humanos
13.
Front Biosci (Landmark Ed) ; 19(8): 1425-35, 2014 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-24896362

RESUMO

ABC-transporters prevent the access of antiepileptic drugs into brain parenchyma, which partly explains why seizures are frequently refractory to AEDs treatment. Overexpression of ABC-transporters and stem-cell markers including CD34, have been detected in malformations of cortical development (MCD) and brain tumors. ABC-transporters are constitutively expressed during maturation of normal progenitor stem-cells and cancer stem-cells. These abnormal/immature cells of MCD or brain tumors play an active role in the epileptogenesis but the precise nature of this phenomenon is unclear. Irrespective of their property in the pharmacoresistance, ABCB1-transporter P-glycoprotein also plays a role in the membrane depolarization, suggesting that constitutive P-glycprotein overexpression in MCD and brain tumors could explain their epileptogeneic properties. MCD as wells as brain tumors arise from abnormal progenitor cells, where ABC-t together with others stem cell markers, could help to better identification of these abnormal progenitor cells and serve as biomarker of risk for seizure relapse after epilepsy surgery.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Biomarcadores/metabolismo , Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Epilepsia/metabolismo , Células-Tronco/metabolismo , Anticonvulsivantes/uso terapêutico , Encéfalo/patologia , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/patologia , Resistência a Medicamentos , Epilepsia/complicações , Epilepsia/tratamento farmacológico , Humanos , Malformações do Desenvolvimento Cortical/metabolismo
14.
Int J Biochem Cell Biol ; 45(7): 1377-88, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23603149

RESUMO

The neuroinflammatory process is thought to contribute to the progression of neurological disorders and brain pathologies. The release of pro-inflammatory cytokines and chemokines by activated glial cells, astrocytes and microglia plays an important role in this process. However, the role of hypoxia-inducible factor-1α (HIF-1α), the key transcription factor regulating the expression of hypoxia-inducible genes, during glial activation is less known. Thus, we examined the significance of HIF-1α in three experimental models: first in an acute model of inflammation induced by pro-inflammatory cytokines TNF-α, IL-1ß and IFN-γ; secondly, in a chronic model of inflammation using an APPswe/PS1dE9 (APP/PS1) transgenic mouse model of Alzheimer's disease and thirdly via the inhibition of the PI3K/AKT pathway in a model of neuronal apoptosis. During acute glial inflammation induced by in vitro administration of TNF-α, IL-1ß and IFN-γ, mRNA expression levels of HIF-1α were significantly upregulated; however, this effect was blocked by SP600126, a pharmacological inhibitor of mitogen-activated protein kinases (MAPKs). These data suggest that MAPKs could be involved in HIF-1α regulation. In addition, we observed that HIF-1α is not involved in the neuronal apoptotic process mediated by PI3-kinase inhibition, which is regulated by c-Jun. Finally, we did not detect significant differences in the expression of HIF-1α mRNA in APP/PS1 mice during the course of the study (3-12 months of age). Thus, we demonstrated that HIF-1α has a prominent role in acute but not in chronic inflammatory processes, such as the one which occurs in the APP/PS1 experimental model of AD. Moreover, HIF-1α is not involved in neuronal apoptosis after PI3K/AKT inhibition.


Assuntos
Doença de Alzheimer/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Animais , Apoptose , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Interferon gama/metabolismo , Interleucina-1beta/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neuroglia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
15.
Drug Metabol Drug Interact ; 26(2): 65-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21756166

RESUMO

Stroke is a major human health problem without efficient available therapeutics. Ischemic brain injury can induce cell death as well as upregulation of endogenous adaptive mechanisms depending on the severity and duration of hypoxia, and the activity of transcription factors, such as hypoxia inducible factor 1-α (HIF-1α). HIF-1α induces gene expression as multidrug resistance (MDR-1) gene associated with drug-refractory phenotype, as well as erythropoietin (Epo) and erythropoietin receptor (Epo-R) associated with O(2) supply. The spontaneous stimulation of the Epo/Epo-R system is not enough for brain protection. Therefore, administration of exogenous recombinant human Epo (rHu-Epo) was suggested as an alternative therapy in stroke. In several experimental models of brain hypoxia, Epo and Epo variants, including rHu-Epo, showed neuroprotective effects. Intranasal administration of these Epo-compounds can reach the central nervous system and protect the brain against ischemia, avoiding hematopoietic effects. However, it has been reported that high expression of Epo-R in neurons must be available to be activated by Epo. According to these considerations, intranasal delivery of rHu-Epo could be an interesting approach in the treatment of cerebral hypoxias avoiding both (i) adverse peripheral effects of treatment with Epo in stroke, and (ii) the pharmacoresistant phenotype depending on MDR-1 expression.


Assuntos
Eritropoetina/administração & dosagem , Hipóxia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/administração & dosagem , Administração Intranasal , Animais , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Receptores da Eritropoetina/análise , Proteínas Recombinantes/administração & dosagem
16.
Neurotox Res ; 20(2): 182-92, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21116766

RESUMO

Stroke is a major human health problem inducing long-term disability without any efficient therapeutic option being currently available. Under hypoxia, hypoxia-inducible factor-1α (HIF-1α) activates several genes as erythropoietin receptor (Epo-R) related with O(2) supply, and the multidrug-resistance gene (MDR-1) related with drug-refractory phenotype. Brain cortical injection of CoCl(2) produces focal hypoxia-like lesion with neuronal and glial alterations, as well as HIF-1α stabilization and MDR-1 overexpression. Intranasal (IN) drug delivery can by-pass blood-brain barrier (BBB) where MDR-1 is normally expressed. We evaluated the effects of IN-rHu-Epo administration on spontaneous motor activity (SMA) and the brain pattern expression of HIF-1α, MDR-1, and Epo-R in our cobalt-induced hypoxia model. Adult male Wistar rats were injected by stereotaxic surgery in frontoparietal cortex, with CoCl(2) (2 µl-50 mM; n = 20) or saline (controls; n = 20). Ten rats of each group were treated with IN-rHu-Epo 24 U or IN-saline. In addition, erythropoietic stimulation was evaluated by reticulocytes (Ret) account during three consecutive days, after intraperitoneal (i.p.)-recombinant-human Epo (rHu-Epo) (950 U; n = 6) or IN-rHu-Epo (24 U; n = 6) administration. SMA was evaluated by open field and rotarod tests, before and after surgical procedures during five consecutive days. Histological and immunostaining studies of HIF-1α, MDR-1, and Epo-R were performed on brain slides. A significant difference in SMA was observed in the hypoxic rats of IN-rHu-Epo-administered group as compared with Co-Saline-treated subjects and controls (p < 0.001). HIF-1α, EPO-R, and MDR-1 were overexpressed in the hypoxic cortex areas, while in contralateral hemisphere or controls, they were negatives. Reticulocytes were only increased in intraperitoneal (i.p.)-rHu-Epo-administered group. In spite of MDR-1 overexpression being detected in neurons, the coexpression of Epo-R could explain the positive effects observed on SMA of IN-rHu-Epo-administered group.


Assuntos
Cobalto/toxicidade , Eritropoetina/administração & dosagem , Comportamento Exploratório/efeitos dos fármacos , Hipóxia Encefálica/induzido quimicamente , Hipóxia Encefálica/tratamento farmacológico , Recuperação de Função Fisiológica/efeitos dos fármacos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Administração Intranasal , Análise de Variância , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Reticulócitos/efeitos dos fármacos , Teste de Desempenho do Rota-Rod
17.
Mol Pharmacol ; 73(5): 1343-6, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18314494

RESUMO

ATP-binding cassette transporters such as P-glycoprotein (Pgp), multidrug resistance-associated protein, and breast cancer resistance protein are known to transport a wide range of substrates and are highly expressed in the capillary endothelial cells that form part of the blood-brain barrier. It is noteworthy that P-glycoprotein has been shown to be up-regulated in animal models of refractory epilepsy, and adding a Pgp inhibitor to treatment regimens has been shown to reverse the drug-resistant phenotype. Limited data have suggested a role for Pgp in epilepsy in humans as well. However, few epilepsy drugs have been shown to be transported by Pgp, leading to controversy over whether Pgp actually plays a role in drug-resistant epilepsy. In this issue of Molecular Pharmacology, Bauer et al. (p. 1444) demonstrate that glutamate can cause localized up-regulation of Pgp via cyclooxygenase-2 (COX-2) and that this phenomenon can be prevented with COX-2 inhibitors. Localized rather than global up-regulation of Pgp may explain some of the difficulty investigators have had in proving a role for Pgp in epilepsy. The results add new support for future clinical trials targeting Pgp expression in drug-refractory epilepsy.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Convulsões/patologia , Animais , Capilares/efeitos dos fármacos , Capilares/enzimologia , Capilares/patologia , Ciclo-Oxigenase 2/metabolismo , Ácido Glutâmico/farmacologia , Camundongos , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/induzido quimicamente , Regulação para Cima/genética
18.
Epilepsia ; 48 Suppl 5: 140-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17910594

RESUMO

It is estimated 20-25% of the epileptic patients fails to achieve good control with the different antiepileptic drugs (AEDs) treatments, developing refractory epilepsy (RE). Discovered first in cancer, the activity of P-glycoprotein (P-gp) and others ABC transporters as multidrug-resistance-associated proteins (MRPs) and breast cancer resistant protein (BCRP) are directly related with the refractoriness. We have observed the overexpression of these all transporters in the brain of patients with RE, and according with other authors, all these data suggests an active drug efflux from brain. Both constitutive and seizure induced brain P-gp overexpression was also suggested. As confirmation of these clinical evidences, different models of experimental epilepsy have demonstrated P-gp overexpression on blood brain barrier (BBB) and brain parenchyma cells, as astrocytes and neurons. In our model, early P-pg detection in vessel-related cells and later additional P-gp detection in neurons, correlated with the gradual loss of protective effect of phenytoin. The progressive neuronal P-gp expression, depending on intensity and time-constancy of seizure-injury, was in agreement with the development of "P-gp-positive seizure-axis" proposed by Kwan & Brodie, who also showed that the development of RE directly correlated with the number and frequency of seizures before initiation of drug therapy. P-gp expression in excretory organs suggests that P-gp have a central role in drug elimination. Persistent low levels of AEDs in plasma and P-gp brain overexpression in several RE pediatric patients were reported. We also observed in adult RE patients, an increased liver clearance of 99mTc-hexakis-2-methoxyisobutylisonitrile (99mTc-MIBI) (a P-gp substrate), and the surgically treated cases showed P-gp brain overexpression. These results suggest the systemic hyperactivity of P-gp in RE patients, including brain P-gp over-expression should be suspected when persistent subtherapeutic levels of AEDs in plasma are detected. P-gp neuronal expression described in both clinical and experimental reports indicates that additional mechanisms could be operative from seizure-affected P-gp-positive neurons, due to AEDs targets are expressed at membrane level. An alternative mechanism was demonstrated in P-gp-expressed cells that exhibit lower membrane potential (Deltapsi(0)=-10 to -20) compared to normal physiological Deltapsi(0) of -60 mV. Under this situation and irrespective to the P-gp pharmacoresistant property or type of drug treatment selected, P-gp-expressed neurons could increase their sensitivity to new seizures perhaps as an epileptogenic mechanism. The understanding of properties of these ABC transporters can offer new tools for better selection of more effective preventive or therapeutic strategies and avoid the invasive surgical treatments for RE.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Anticonvulsivantes/uso terapêutico , Resistência a Múltiplos Medicamentos/fisiologia , Epilepsia/tratamento farmacológico , Epilepsia/fisiopatologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/farmacologia , Animais , Anticonvulsivantes/sangue , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Resistência a Múltiplos Medicamentos/genética , Epilepsia/genética , Expressão Gênica/genética , Humanos , Fígado/metabolismo , Taxa de Depuração Metabólica , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas de Neoplasias/genética , Nitrilas/metabolismo , Compostos de Organotecnécio/metabolismo , Ratos , Distribuição Tecidual
19.
J Neurol Sci ; 258(1-2): 84-92, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17459414

RESUMO

Neuronal damage after stroke-associated brain hypoxia is a leading cause of long-term disability and death. The refractoriness to therapeutic strategies for neuroprotection after 3 h post brain ischemia is poorly understood. P-glycoprotein (P-gp), the multidrug resistance gene (MDR-1) product is normally expressed at blood-brain-barrier. P-gp neuronal expression has been demonstrated in refractory epilepsy and after brain ischemia. In this report we investigated the hypoxia-induced neuronal P-gp expression after local injection of CoCl(2) (1-200 mM) in the fronto-parietal cortex of male adult rats (Bregma -1.30 mm) by stereotaxic surgery. P-gp immunostaining of brain slides was analyzed using specific monoclonal antibodies and double immunolabeling was done with specific astrocytic and neuronal markers. Five days after injection of 1 mM CoCl(2), P-gp expression surrounding the lesion site was observed in neurons, astrocytic end-foot on capillary blood vessels and endothelial cells on blood vessels. Higher CoCl(2) doses (200 mM) resulted in additional P-gp immunostaining of the entire astrocytic and neuronal cytoplasm. Electron microscopy (EM) studies showed alterations in neurons as early as 6 h after the CoCl(2) injection. P-gp expression in hypoxic neurons and astrocytic end-foot could potentially impair of drugs access to the brain parenchyma thus suggesting the presence of two P-gp-based pumping systems (one in astrocytes and other in the hypoxic neurons) that are able to behave as a previously unnoticed obstacle for pharmacological strategies of neuroprotection.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Expressão Gênica/fisiologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Neurônios/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Antimutagênicos/administração & dosagem , Cobalto/administração & dosagem , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Hipóxia/induzido quimicamente , Hipóxia/patologia , Masculino , Microscopia Eletrônica de Transmissão/métodos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Lobo Parietal/efeitos dos fármacos , Lobo Parietal/metabolismo , Lobo Parietal/patologia , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Wistar
20.
Hematología (B. Aires) ; 11(1): 14-19, ene.-abr. 2007. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-526632

RESUMO

El tratamiento de la Leucemia Mieloide Crónica (LMC) con Imatinib puede fracasar por mutaciones en el dominio tirosina quinasa o amplificación del gen BCR/ABL. Otros mecanismos de refractariedad pueden deberse a los genes de resistencia múltiple a drogas (MDR). Objetivo: Investigar en la línea celular K562 (LMC resistente al Imatinib), la expresión del gen MDRl y los efectos de su inhibición con Ciclosporina A (CyA). Métodos: Se sintetizó ADNc por retrotranscripción del ARN total y se amplificaron por RT-PCR los genes BCR/ABL y MDRl con primers específicos. Se verificó la expresión de la glicoproteína P-gp (producto del gen MDRl) por inmunohistoquímica con 2 anticuerpos monoclonales (C494 y C2l9). Las células K562 fueron enfrentadas (24hs) con Imatinib (2uM), CyA (3ug/ml), Imatinib+Cy A. Se evaluaron apoptosis con naranja de acridina/bromuro de etidio (microscopía de fluorescencia) y función farmacorresistente de P-gp con Rhodamina-l23 (citometría de flujo). Resultados: La expresión del gen MDRl se confirmó tanto por RT-PCRcomo por inmunhistoquímica. La prueba funcional con Rhodamina-l23 indicó que la P-gp fue inhibida por CyA o hipotermia. El tratamiento con Imatinib+Cy A triplicó el porcentaje de apoptosis comparado con Imatinib solo. Conclusión: El gen MDRl jugaría un rol adicional en la resistencia al Imatinib. La Cy A u otros inhibido res de la P-gp, facilitaría la acción del Imatinib, induciendo mayor porcentaje de apoptosis en células BCR/ ABL positivas.


Assuntos
Genes MDR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA