Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Nature ; 618(7963): 159-168, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37225977

RESUMO

Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.


Assuntos
Regeneração Nervosa , Humanos , Neoplasias/tratamento farmacológico , Regeneração Nervosa/efeitos dos fármacos , Isoformas de Proteínas/agonistas , Transdução de Sinais/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/química , Classe I de Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Cardiotônicos/farmacologia , Animais , Biocatálise/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Compressão Nervosa , Proliferação de Células/efeitos dos fármacos
2.
Cell Physiol Biochem ; 55(S3): 108-130, 2021 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-34043299

RESUMO

Transient receptor potential vanilloid (TRPV) channels are part of the TRP channel superfamily and named after the first identified member TRPV1, that is sensitive to the vanillylamide capsaicin. Their overall structure is similar to the structure of voltage gated potassium channels (Kv) built up as homotetramers from subunits with six transmembrane helices (S1-S6). Six TRPV channel subtypes (TRPV1-6) are known, that can be subdivided into the thermoTRPV (TRPV1-4) and the Ca2+-selective TRPV channels (TRPV5, TRPV6). Contrary to Kv channels, TRPV channels are not primary voltage gated. All six channels have distinct properties and react to several endogenous ligands as well as different gating stimuli such as heat, pH, mechanical stress, or osmotic changes. Their physiological functions are highly diverse and subtype as well as tissue specific. In many tissues they serve as sensors for different pain stimuli (heat, pressure, pH) and contribute to the homeostasis of electrolytes, the maintenance of barrier functions and the development of macrophages. Due to their fundamental role in manifold physiological and pathophysiological processes, TRPV channels are promising targets for drug development. However, drugs targeting specific TRPV channels, that are suitable for drug therapy, are rare. Moreover, selective and potent compounds for further research at TRPV channels are often lacking. In this review different aspects of the structure, the different gating stimuli, the expression pattern, the physiological and pathophysiological roles as well as the modulating mechanisms of synthetic, natural and endogenous ligands are summarized.


Assuntos
Analgésicos/farmacologia , Antineoplásicos/farmacologia , Fatores Imunológicos/farmacologia , Moduladores de Transporte de Membrana/farmacologia , Canais de Cátion TRPV/metabolismo , Analgésicos/química , Analgésicos/classificação , Antineoplásicos/química , Antineoplásicos/classificação , Sítios de Ligação , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Humanos , Fatores Imunológicos/química , Fatores Imunológicos/classificação , Ativação do Canal Iônico/efeitos dos fármacos , Ligantes , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Moduladores de Transporte de Membrana/química , Moduladores de Transporte de Membrana/classificação , Modelos Moleculares , Especificidade de Órgãos , Ligação Proteica , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/classificação , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/classificação
3.
J Med Chem ; 64(8): 5123-5136, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33793232

RESUMO

The retinoid X receptors (RXR) are ligand-activated transcription factors involved in multiple regulatory networks as universal heterodimer partners for nuclear receptors. Despite their high therapeutic potential in many pathologies, targeting of RXR has only been exploited in cancer treatment as the currently available RXR agonists suffer from exceptional lipophilicity, poor pharmacokinetics (PK), and adverse effects. Aiming to overcome the limitations and to provide improved RXR ligands, we developed a new potent RXR ligand chemotype based on the nonsteroidal anti-inflammatory drug oxaprozin. Systematic structure-activity relationship analysis enabled structural optimization toward low nanomolar potency similar to the well-established rexinoids. Cocrystal structures of the most active derivatives demonstrated orthosteric binding, and in vivo profiling revealed superior PK properties compared to current RXR agonists. The optimized compounds were highly selective for RXR activation and induced RXR-regulated gene expression in native cellular and in vivo settings suggesting them as excellent chemical tools to further explore the therapeutic potential of RXR.


Assuntos
Oxaprozina/análogos & derivados , Receptores X de Retinoides/agonistas , Animais , Sítios de Ligação , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Meia-Vida , Humanos , Ligantes , Camundongos , Microssomos/metabolismo , Simulação de Dinâmica Molecular , Oxaprozina/metabolismo , Oxaprozina/farmacologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pirazóis/química , Pirazóis/metabolismo , Pirazóis/farmacologia , Ratos , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Relação Estrutura-Atividade
4.
Front Immunol ; 12: 783780, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35003101

RESUMO

Cells are richly equipped with nuclear receptors, which act as ligand-regulated transcription factors. Peroxisome proliferator activated receptors (PPARs), members of the nuclear receptor family, have been extensively studied for their roles in development, differentiation, and homeostatic processes. In the recent past, there has been substantial interest in understanding and defining the functions of PPARs and their agonists in regulating innate and adaptive immune responses as well as their pharmacologic potential in combating acute and chronic inflammatory disease. In this review, we focus on emerging evidence of the potential roles of the PPAR subtypes in macrophage biology. We also discuss the roles of dual and pan PPAR agonists as modulators of immune cell function, microbial infection, and inflammatory diseases.


Assuntos
Regulação da Expressão Gênica/imunologia , Fatores Imunológicos/farmacologia , Macrófagos/imunologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/genética , Animais , Modelos Animais de Doenças , Retroalimentação Fisiológica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Fatores Imunológicos/uso terapêutico , Infecções/tratamento farmacológico , Infecções/imunologia , Infecções/microbiologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Receptores Ativados por Proliferador de Peroxissomo/genética , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
Chem Biol Drug Des ; 97(6): 1129-1136, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33283479

RESUMO

Human transcriptional enhanced associate domain (TEAD) family consists of four paralogous transcription factors that function to modulate gene expression by interacting with YAP-like coactivators and have been recognized as potential therapeutic targets of diverse diseases including lung cancer and gastric tumor. Here, we attempt to explore the systematic interaction profile between the 4 TEAD proteins and the peptides derived from the binding sites of 8 known YAP-like coactivators, in order to analyze the binding affinity and recognition specificity of these peptides toward the TEAD family, and to design hydrocarbon-stapled/cyclized peptides that can target the specific interaction profile for each coactivator. Structural, energetic, and dynamic investigations of TEAD-coactivator interactions reveal that the coactivators adopt three independent secondary structure regions (ß-strand, α-helix, and Ω-loop) to surround on the surface of TEAD proteins, in which the α-helical and Ω-loop regions are primarily responsible for the interactions. Five α-helical peptides and four Ω-loop peptides are derived from the 8 YAP-like coactivators, and their systematic binding profile toward the 4 TEAD proteins is created, and hydrocarbon stapling and cyclization strategies are employed to constrain the free α-helical and Ω-loop peptides into their native conformations, respectively, thus effectively promoting peptide binding to TEADs. The all-hydrocarbon and disulfide bridges are designed to point out the TEAD-peptide complex interface, which would not disrupt the direct intermolecular interaction between the TEAD and peptide. Therefore, the stapling and cyclization only improve peptide binding affinity to these TEADs, but do not alter peptide recognition specificity over different TEADs.


Assuntos
Proteínas de Ciclo Celular/química , Hidrocarbonetos/química , Peptídeos/química , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Ciclização , Polarização de Fluorescência , Humanos , Simulação de Dinâmica Molecular , Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Fatores de Transcrição/agonistas , Fatores de Transcrição/química
6.
Bioorg Med Chem Lett ; 30(17): 127391, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32738999

RESUMO

Nonpeptide sst2 agonists can provide a new treatment option for patients with acromegaly, carcinoid tumors, and neuroendocrine tumors. Our medicinal chemistry efforts have led to the discovery of novel 3,4-dihydroquinazoline-4-carboxamides as sst2 agonists. This class of molecules exhibits excellent human sst2 potency and selectivity against sst1, sst3, sst4 and sst5 receptors. Leading compound 3-(3-chloro-5-methylphenyl)-6-(3-fluoro-2-hydroxyphenyl)-N,7-dimethyl-N-{[(2S)-pyrrolidin-2-yl]methyl}-3,4-dihydroquinazoline-4-carboxamide (28) showed no inhibition of major CYP450 enzymes (2C9, 2C19, 2D6 and 3A4) and weak inhibition of the hERG channel.


Assuntos
Amidas/química , Receptores de Somatostatina/agonistas , Amidas/metabolismo , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Receptores de Somatostatina/metabolismo , Relação Estrutura-Atividade
7.
Sci Rep ; 10(1): 2990, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32076076

RESUMO

Photoreceptor cell death is the ultimate cause of vision loss in many retinal disorders, and there is an unmet need for neuroprotective modalities to improve photoreceptor survival. Similar to cancer cells, photoreceptors maintain pyruvate kinase muscle isoform 2 (PKM2) expression, which is a critical regulator in aerobic glycolysis. Unlike PKM1, which has constitutively high catalytic activity, PKM2 is under complex regulation. Recently, we demonstrated that genetically reprogramming photoreceptor metabolism via PKM2-to-PKM1 substitution is a promising neuroprotective strategy. Here, we explored the neuroprotective effects of pharmacologically activating PKM2 via ML-265, a small molecule activator of PKM2, during acute outer retinal stress. We found that ML-265 increased PKM2 activity in 661 W cells and in vivo in rat eyes without affecting the expression of genes involved in glucose metabolism. ML-265 treatment did, however, alter metabolic intermediates of glucose metabolism and those necessary for biosynthesis in cultured cells. Long-term exposure to ML-265 did not result in decreased photoreceptor function or survival under baseline conditions. Notably, though, ML-265-treatment did reduce entrance into the apoptotic cascade in in vitro and in vivo models of outer retinal stress. These data suggest that reprogramming metabolism via activation of PKM2 is a novel, and promising, therapeutic strategy for photoreceptor neuroprotection.


Assuntos
Apoptose/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Células Fotorreceptoras/efeitos dos fármacos , Piridazinas/farmacologia , Pirróis/farmacologia , Piruvato Quinase/metabolismo , Doenças Retinianas/tratamento farmacológico , Animais , Cegueira/etiologia , Cegueira/prevenção & controle , Linhagem Celular , Modelos Animais de Doenças , Ativadores de Enzimas/uso terapêutico , Glicólise/efeitos dos fármacos , Humanos , Injeções Intravítreas , Masculino , Camundongos , Camundongos Knockout , Células Fotorreceptoras/patologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Piridazinas/uso terapêutico , Pirróis/uso terapêutico , Piruvato Quinase/genética , Coelhos , Ratos , Doenças Retinianas/complicações , Doenças Retinianas/patologia
8.
J Med Chem ; 62(19): 8711-8732, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31532644

RESUMO

Clinical development of catechol-based orthosteric agonists of the dopamine D1 receptor has thus far been unsuccessful due to multiple challenges. To address these issues, we identified LY3154207 (3) as a novel, potent, and subtype selective human D1 positive allosteric modulator (PAM) with minimal allosteric agonist activity. Conformational studies showed LY3154207 adopts an unusual boat conformation, and a binding pose with the human D1 receptor was proposed based on this observation. In contrast to orthosteric agonists, LY3154207 showed a distinct pharmacological profile without a bell-shaped dose-response relationship or tachyphylaxis in preclinical models. Identification of a crystalline form of free LY3154207 from the discovery lots was not successful. Instead, a novel cocrystal form with superior solubility was discovered and determined to be suitable for development. This cocrystal form was advanced to clinical development as a potential first-in-class D1 PAM and is now in phase 2 studies for Lewy body dementia.


Assuntos
Isoquinolinas/farmacologia , Receptores de Dopamina D1/agonistas , Acetilcolina/metabolismo , Administração Oral , Regulação Alostérica/efeitos dos fármacos , Animais , Sítios de Ligação , Cristalografia por Raios X , AMP Cíclico/metabolismo , Células HEK293 , Meia-Vida , Humanos , Isoquinolinas/química , Isoquinolinas/farmacocinética , Rim/efeitos dos fármacos , Rim/metabolismo , Locomoção/efeitos dos fármacos , Camundongos , Conformação Molecular , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Ratos , Receptores de Dopamina D1/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
9.
Cell Mol Life Sci ; 76(20): 3969-3985, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31218451

RESUMO

Cardiovascular diseases represent one of the most important health problems of developed countries. One of the main actors involved in the onset and development of cardiovascular diseases is the increased production of reactive oxygen species that, through lipid peroxidation, protein oxidation and DNA damage, induce oxidative stress and cell death. Basic and clinical research are ongoing to better understand the endogenous antioxidant mechanisms that counteract oxidative stress, which may allow to identify a possible therapeutic targeting/application in the field of stress-dependent cardiovascular pathologies. In this context, increasing attention is paid to the glutathione/glutathione-peroxidase and to the thioredoxin/thioredoxin-reductase systems, among the most potent endogenous antioxidative systems. These key enzymes, belonging to the selenoprotein family, have a well-established function in the regulation of the oxidative cell balance. The aim of the present review was to highlight the role of selenoproteins in cardiovascular diseases, introducing the emerging cardioprotective role of endoplasmic reticulum-resident members and in particular one of them, namely selenoprotein T or SELENOT. Accumulating evidence indicates that the dysfunction of different selenoproteins is involved in the susceptibility to oxidative stress and its associated cardiovascular alterations, such as congestive heart failure, coronary diseases, impaired cardiac structure and function. Some of them are under investigation as useful pathological biomarkers. In addition, SELENOT exhibited intriguing cardioprotective effects by reducing the cardiac ischemic damage, in terms of infarct size and performance. In conclusion, selenoproteins could represent valuable targets to treat and diagnose cardiovascular diseases secondary to oxidative stress, opening a new avenue in the field of related therapeutic strategies.


Assuntos
Cardiotônicos/uso terapêutico , Doenças Cardiovasculares/genética , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Peptídeos/uso terapêutico , Selenocisteína/metabolismo , Selenoproteínas/genética , Animais , Antioxidantes/metabolismo , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/terapia , Regulação da Expressão Gênica , Glutationa Peroxidase/metabolismo , Humanos , Terapia de Alvo Molecular/métodos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Selenoproteínas/agonistas , Selenoproteínas/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo
10.
J Steroid Biochem Mol Biol ; 191: 105312, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30995525

RESUMO

ERbeta (ERß) celebrated its 20th birthday in 2016 and although the overwhelming data in the literature indicate a role for this receptor in the control of epithelial proliferation, neurodegeneration and immune function, no ERß agonists have yet made it to the clinics. This is the situation, despite the fact that very good safe ERß agonists have been synthesized and at least one has been donated to the NIH for distribution to researchers, who want to study its possible clinical use. Clinical trials are ongoing for the use of ERß agonists in prostate cancer and schizophrenia but even today reviewers of our grants still make comments like "The grant is excellent except that the focus of the grant is ERß". There are multiple reasons for the non-acceptance of the value of ERß and in this paper we will discuss issues raised by labs which do not support a role for ERß in physiology or pathology.


Assuntos
Receptor beta de Estrogênio/metabolismo , Animais , Anticorpos/imunologia , Descoberta de Drogas , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/imunologia , Expressão Gênica , Humanos , Imunidade , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo
11.
Steroids ; 142: 6-13, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-28962850

RESUMO

The role played by progestogens in modulating Schwann cell pathophysiology is well established. Progestogens exert their effects in these cells through both classical genomic and non-genomic mechanisms, the latter mediated by the GABA-A receptor. However, there is evidence that other receptors may be involved. Membrane progesterone receptors (mPRs) are novel 7-transmembrane receptors coupled to G proteins that have been characterized in different tissues and cells, including the central nervous system (CNS). The mPRs were shown to mediate some of progestogens' neuroprotective effects in the CNS, and to be upregulated in glial cells after traumatic brain injury. Based on this evidence, this paper investigated the possible involvement of mPRs in mediating progestogen actions in S42 Schwann cells. All five mPR isoforms and progesterone receptor membrane component 1 (PGRMC1) were detected in Schwann cells, and were present on the cell membrane. Progesterone and the mPR-specific agonist, Org-OD-02-0 (02) bound to these membranes, indicating the presence of functional mPRs. The mPR agonist 02 rapidly increased cell migration in an in vitro assay, suggesting a putative role of mPRs in the nerve regeneration process. Treatment with pertussis toxin and 8-Br-cAMP blocked 02-induced cell migration, suggesting this progestogen action is mediated by activation of an inhibitory G protein, leading to a decrease in intracellular cAMP levels. In contrast, long-term mPR activation led to increased expression levels of myelin associated glycoprotein (MAG). Taken together, these findings show that mPRs are present and active in Schwann cells and have a role in modulating their physiological processes.


Assuntos
Membrana Celular/metabolismo , Movimento Celular , Glicoproteína Associada a Mielina/biossíntese , Neuroglia/citologia , Neuroglia/metabolismo , Receptores de Progesterona/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Neuroglia/efeitos dos fármacos , Toxina Pertussis/farmacologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/análise , Isoformas de Proteínas/metabolismo , Ratos , Receptores de Progesterona/agonistas , Receptores de Progesterona/análise , Tionucleotídeos/farmacologia , Células Tumorais Cultivadas
12.
Int J Mol Sci ; 19(8)2018 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-30060458

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are a well-known pharmacological target for the treatment of multiple diseases, including diabetes mellitus, dyslipidemia, cardiovascular diseases and even primary biliary cholangitis, gout, cancer, Alzheimer's disease and ulcerative colitis. The three PPAR isoforms (α, ß/δ and γ) have emerged as integrators of glucose and lipid metabolic signaling networks. Typically, PPARα is activated by fibrates, which are commonly used therapeutic agents in the treatment of dyslipidemia. The pharmacological activators of PPARγ include thiazolidinediones (TZDs), which are insulin sensitizers used in the treatment of type 2 diabetes mellitus (T2DM), despite some drawbacks. In this review, we summarize 84 types of PPAR synthetic ligands introduced to date for the treatment of metabolic and other diseases and provide a comprehensive analysis of the current applications and problems of these ligands in clinical drug discovery and development.


Assuntos
Descoberta de Drogas , Doenças Metabólicas/tratamento farmacológico , Terapia de Alvo Molecular , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Receptores Ativados por Proliferador de Peroxissomo/química , Animais , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças , Humanos , Hipolipemiantes/uso terapêutico , Ligantes , Camundongos , Conformação Molecular , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratos
13.
Endocrine ; 62(2): 477-486, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30027434

RESUMO

PURPOSE: The importance of androgen receptor variants (AR-Vs) is recognized in prostate cancer. AR-Vs have been the focus of many studies. Expression of AR-Vs has been proposed as a biomarker for resistance to androgen deprivation therapy for metastatic disease. Herein, we show dynamic changes in AR-Vs expression in response to androgen modulation. METHODS: The C4-2B cell line was exposed to low (10-13 M) and high (10-8 M) androgen (dihydrotestosterone, DHT) levels, with or without flutamide. mRNA and protein expression levels were assessed by qPCR and immunohistochemistry, respectively. RESULTS: We demonstrated that high levels of DHT downregulate AR-FL and AR-Vs. Even though AR-Vs did not present ligand-binding domain, thus were not capable of binding to DHT, they present dynamic changes under androgen treatment. Treatment with flutamide alone or in association with low levels of DHT stimulates growth of prostatic cells. CONCLUSIONS: Importantly, we provide evidence that AR-Vs respond differently to androgenic modulation. These findings have implications for a better understanding of the role of AR-Vs in prostate carcinogenesis.


Assuntos
Androgênios/farmacologia , Proteínas Mutantes , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Masculino , Proteínas Mutantes/agonistas , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Polimorfismo Genético , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-29698686

RESUMO

In the present study, we cloned and characterized two somatostatin (SS) receptors (SSTRs) from topmouth culter (Erythroculter ilishaeformis) designated as EISSTR6 and EISSTR7. Analysis of EISSTR6 and EISSTR7 signature motifs, 3D structures, and homology with the known members of the SSTR family indicated that the novel receptors had high similarity to the SSTRs of other vertebrates. EISSTR6 and EISSTR7 mRNA expression was detected in 17 topmouth culter tissues, and the highest level was observed in the pituitary. Luciferase reporter assay revealed that SS14 significantly inhibited forskolin-stimulated pCRE-luc promoter activity in HEK293 cells transiently expressing EISSTR6 and EISSTR7, indicating that the receptors can be activated by SS14. We also identified phosphorylation sites important for the functional activity of EISSTR6 and EISSTR7 by mutating Ser23, 43, 107, 196, 311 and Ser7, 29, 61, 222, 225 residues, respectively, to Ala, which significantly reduced the inhibitory effects of SS14 on the CRE promoter mediated by EISSTR6 and EISSTR7. Furthermore, treatment of juvenile topmouth culters with microcystin-LR or 17ß-estradiol significantly affected EISSTR6 and EISSTR7 transcription in the brain, liver and spleen, suggesting that these receptors may be involved in the pathogenic mechanisms induced by endocrine disruptors. Our findings should contribute to the understanding of the structure-function relationship and evolution of the SSTR family.


Assuntos
Cyprinidae/metabolismo , Proteínas de Peixes/metabolismo , Modelos Moleculares , Hipófise/metabolismo , Receptores de Somatostatina/metabolismo , Motivos de Aminoácidos , Animais , Bases de Dados de Proteínas , Disruptores Endócrinos/toxicidade , Feminino , Proteínas de Peixes/agonistas , Proteínas de Peixes/química , Proteínas de Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células HEK293 , Humanos , Especificidade de Órgãos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Filogenia , Hipófise/efeitos dos fármacos , Conformação Proteica , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Somatostatina/química , Somatostatina/metabolismo
15.
Cell Death Dis ; 9(5): 500, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29703889

RESUMO

Necroptosis, a form of regulated necrotic cell death, is mediated by receptor interacting protein 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like protein (MLKL). However, the mechanism by which necroptosis promotes inflammation is still unclear. Here we report that the expression of cytokines is robustly upregulated in a cell-autonomous manner during necroptosis induced by tumor necrosis factor alpha (TNFα). We demonstrate that TNFα-induced necroptosis leads to two waves of cytokine production. The first wave, more transient and weaker than the second, is in response to TNFα alone; whereas the second wave depends upon the necroptotic signaling. We show that necroptosis promotes the transcription of TNFα-target genes in a cell-intrinsic manner. The activation of both NF-κB and p38 by the necroptotic machinery, RIPK1, RIPK3, and MLKL, is involved in mediating the robust induction of cytokine expression in the second wave. In contrast, necroptosis induced by direct oligomerization of MLKL promotes cytokine production at much lower levels than that of necroptosis induced with TNFα. Thus, we conclude that TNFα-induced necroptosis signaling events mediated by RIPK1 and RIPK3 activation, in addition to the MLKL oligomerization, promotes the expression of cytokines involving multiple intracellular signaling mechanisms including NF-κB pathway and p38. These findings reveal that the necroptotic cell death machinery mounts an immune response by promoting cell-autonomous production of cytokines. Our study provides insights into the mechanism by which necroptosis promotes inflammation in human diseases.


Assuntos
Quimiocina CXCL1/genética , Regulação da Expressão Gênica/imunologia , Necrose/genética , Proteínas Quinases/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/imunologia , Linhagem Celular , Quimiocina CXCL1/agonistas , Quimiocina CXCL1/imunologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Células HEK293 , Células HT29 , Humanos , Camundongos , Necrose/imunologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteínas Quinases/imunologia , Multimerização Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Transdução de Sinais , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
16.
CNS Neurosci Ther ; 24(9): 763-774, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29691988

RESUMO

While the insulin receptor (IR) was found in the CNS decades ago, the brain was long considered to be an insulin-insensitive organ. This view is currently revisited, given emerging evidence of critical roles of IR-mediated signaling in development, neuroprotection, metabolism, and plasticity in the brain. These diverse cellular and physiological IR activities are distinct from metabolic IR functions in peripheral tissues, thus highlighting region specificity of IR properties. This particularly concerns the fact that two IR isoforms, A and B, are predominantly expressed in either the brain or peripheral tissues, respectively, and neurons express exclusively IR-A. Intriguingly, in comparison with IR-B, IR-A displays high binding affinity and is also activated by low concentrations of insulin-like growth factor-2 (IGF-2), a regulator of neuronal plasticity, whose dysregulation is associated with neuropathologic processes. Deficiencies in IR activation, insulin availability, and downstream IR-related mechanisms may result in aberrant IR-mediated functions and, subsequently, a broad range of brain disorders, including neurodevelopmental syndromes, neoplasms, neurodegenerative conditions, and depression. Here, we discuss findings on the brain-specific features of IR-mediated signaling with focus on mechanisms of primary receptor activation and their roles in the neuropathology. We aimed to uncover the remaining gaps in current knowledge on IR physiology and highlight new therapies targeting IR, such as IR sensitizers.


Assuntos
Antígenos CD/metabolismo , Encéfalo/metabolismo , Doenças do Sistema Nervoso Central/tratamento farmacológico , Doenças do Sistema Nervoso Central/metabolismo , Receptor de Insulina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Doenças do Sistema Nervoso Central/patologia , Humanos , Insulina/administração & dosagem , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Receptor de Insulina/agonistas , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Resultado do Tratamento
17.
Vitam Horm ; 107: 27-66, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29544634

RESUMO

Gonadotropin-releasing hormone (GnRH) is recognized as the central regulator of the functions of the pituitary-gonadal axis. The increasing knowledge on the mechanisms controlling the development and the function of GnRH-producing neurons is leading to a better diagnostic and therapeutic approach for hypogonadotropic hypogonadisms and for alterations of the puberty onset. During female life span, the function of the GnRH pulse generator may be affected by a number of inputs from other neuronal systems, offering alternative strategies for diagnostic and therapeutic interventions. Moreover, the identification of a GnRH/GnRH receptor system in both human ovary and endometrium has widened the spectrum of action of the peptide outside its hypothalamic functions. The pharmacological use of GnRH itself or its synthetic analogs (agonists and antagonists) provides a valid tool to either stimulate or block gonadotropin secretion and to modulate the female fertility in several reproductive disorders and in assisted reproduction technology. The use of GnRH agonists in young female patients undergoing chemotherapy is also considered a promising therapeutic approach to counteract iatrogenic ovarian failure.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Modelos Biológicos , Células Neuroendócrinas/fisiologia , Ovário/fisiologia , Precursores de Proteínas/metabolismo , Receptores LHRH/agonistas , Reprodução , Animais , Endométrio/efeitos dos fármacos , Endométrio/crescimento & desenvolvimento , Endométrio/fisiologia , Endométrio/fisiopatologia , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Fármacos para a Fertilidade Feminina/uso terapêutico , Preservação da Fertilidade/tendências , Hormônio Liberador de Gonadotropina/química , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistema Hipotálamo-Hipofisário/fisiopatologia , Infertilidade Feminina/tratamento farmacológico , Infertilidade Feminina/patologia , Infertilidade Feminina/fisiopatologia , Infertilidade Feminina/terapia , Ciclo Menstrual/efeitos dos fármacos , Células Neuroendócrinas/citologia , Células Neuroendócrinas/metabolismo , Células Neuroendócrinas/patologia , Ovário/efeitos dos fármacos , Ovário/crescimento & desenvolvimento , Ovário/fisiopatologia , Gravidez , Insuficiência Ovariana Primária/tratamento farmacológico , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/fisiopatologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/química , Puberdade/efeitos dos fármacos , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/metabolismo , Receptores LHRH/uso terapêutico , Reprodução/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
18.
J Reprod Dev ; 64(2): 101-108, 2018 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-29276208

RESUMO

Oviductal glycoprotein 1 (OVGP1), an oviductin, is involved in the maintenance of sperm viability and motility and contributes to sperm capacitation in the oviduct. In this study, the regulatory effects exerted by prostaglandin E2 (PGE2) and F2α (PGF2α) on OVGP1 expression via their corresponding receptors in bovine oviductal epithelial cells (BOECs) were investigated. BOECs were cultured in vitro, and their expression of receptors of PGE2 (PTGER1, PTGER2, PTGER3, and PTGER4) and PGF2α (PTGFR) was measured using RT-qPCR. Ca2+ concentration was determined with a fluorescence-based method and cAMP was quantified by enzyme-linked immunosorbent assays to verify activation of PTGER2 and PTGFR by their corresponding agonists in these cells. OVGP1 mRNA and protein expression was measured using RT-qPCR and western blotting, respectively, following PTGER2 and PTGFR agonist-induced activation. PTGER1, PTGER2, PTGER4, and PTGFR were found to be present in BOECs; however, PTGER3 expression was not detected. OVGP1 expression was significantly promoted by 10-6 M butaprost (a PTGER2 agonist) and decreased by 10-6 M fluprostenol (a PTGFR agonist). In addition, 3 µM H-89 (a PKA inhibitor) and 3 µM U0126 (an ERK inhibitor) effectively inhibited PGE2-induced upregulation of OVGP1, and 5 µM chelerythrine chloride (a PKC inhibitor) and 3 µM U0126 negated OVGP1 downregulation by PGF2α. In conclusion, this study demonstrates that OVGP1 expression in BOECs is enhanced by PGE2 via PTGER2-cAMP-PKA signaling, and reduced by PGF2α through the PTGFR-Ca2+-PKC pathway.


Assuntos
Dinoprosta/metabolismo , Dinoprostona/metabolismo , Regulação da Expressão Gênica , Glicoproteínas/metabolismo , Oviductos/metabolismo , Receptores de Prostaglandina E Subtipo EP2/agonistas , Receptores de Prostaglandina/agonistas , Matadouros , Alprostadil/análogos & derivados , Alprostadil/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Bovinos , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/agonistas , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Oviductos/citologia , Oviductos/efeitos dos fármacos , Prostaglandinas F Sintéticas/farmacologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo
19.
Int J Mol Sci ; 18(10)2017 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-28934113

RESUMO

Allogeneic haemopoietic stem cell transplantation (HSCT) is increasingly used to treat haematological malignant diseases via the graft-versus-leukaemia (GvL) or graft-versus-tumour effects. Although improvements in infectious disease prophylaxis, immunosuppressive treatments, supportive care, and molecular based tissue typing have contributed to enhanced outcomes, acute graft-versus-host disease and other transplant related complications still contribute to high mortality and significantly limit the more widespread use of HSCT. Sphingosine 1-phosphate (S1P) is a zwitterionic lysophospholipid that has been implicated as a crucial signaling regulator in many physiological and pathophysiological processes including multiple cell types such as macrophages, dendritic cells, T cells, T regulatory cells and endothelial cells. Recent data suggested important roles for S1P signaling in engraftment, graft-versus-host disease (GvHD), GvL and other processes that occur during and after HSCT. Based on such data, pharmacological intervention via S1P modulation may have the potential to improve patient outcome by regulating GvHD and enhancing engraftment while permitting effective GvL.


Assuntos
Azetidinas/farmacologia , Compostos de Benzil/farmacologia , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Lisofosfolipídeos/imunologia , Organofosfatos/farmacologia , Transdução de Sinais/imunologia , Esfingosina/análogos & derivados , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Doença Enxerto-Hospedeiro/patologia , Efeito Enxerto vs Leucemia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Lisofosfolipídeos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/imunologia , Esfingosina/metabolismo , Esfingosina/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transplante Homólogo
20.
Biochemistry ; 56(41): 5471-5475, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-28845660

RESUMO

Chemical biosensors, for which chemical detection triggers a fluorescent signal, have the potential to accelerate the screening of noncolorimetric chemicals produced by microbes, enabling the high-throughput engineering of enzymes and metabolic pathways. Here, we engineer a G-protein-coupled receptor (GPCR)-based sensor to detect serotonin produced by a producer microbe in the producer microbe's supernatant. Detecting a chemical in the producer microbe's supernatant is nontrivial because of the number of other metabolites and proteins present that could interfere with sensor performance. We validate the two-cell screening system for medium-throughput applications, opening the door to the rapid engineering of microbes for the increased production of serotonin. We focus on serotonin detection as serotonin levels limit the microbial production of hydroxystrictosidine, a modified alkaloid that could accelerate the semisynthesis of camptothecin-derived anticancer pharmaceuticals. This work shows the ease of generating GPCR-based chemical sensors and their ability to detect specific chemicals in complex aqueous solutions, such as microbial spent medium. In addition, this work sets the stage for the rapid engineering of serotonin-producing microbes.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Receptores 5-HT4 de Serotonina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Serotonina/análise , Engenharia Celular , Meios de Cultivo Condicionados/química , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Proteínas Ativadoras de GTPase/genética , Galactose/metabolismo , Deleção de Genes , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinética , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Fator de Acasalamento/genética , Receptores 5-HT4 de Serotonina/química , Receptores 5-HT4 de Serotonina/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/agonistas , Proteínas de Saccharomyces cerevisiae/genética , Serotonina/metabolismo , Espectrometria de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA