Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.337
Filtrar
1.
Future Med Chem ; 15(5): 453-465, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-37013895

RESUMO

HIPK2 is a serine/threonine kinase, located in the nucleus, that was initially found to be able to phosphorylate p53 at Ser46 to promote apoptosis; it has been widely studied. It has been reported that HIPK2 can simultaneously regulate TGF-ß/Smad3, Wnt/ß-catenin, Notch and NF-κB pathways in the kidney to initiate inflammation and fibrosis, resulting in the development of chronic kidney disease (CKD). Therefore, inhibition of HIPK2 is strongly considered an effective method for the treatment of CKD. In brief, this review summarizes the progress of HIPK2 in CKD as well as the reported HIPK2 inhibitors and their role in different CKD models.


Assuntos
Proteínas de Transporte , Proteínas Serina-Treonina Quinases , Insuficiência Renal Crônica , Humanos , Apoptose , Proteínas de Transporte/antagonistas & inibidores , Fibrose , Rim , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Insuficiência Renal Crônica/patologia
2.
Eur J Med Chem ; 236: 114311, 2022 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-35385803

RESUMO

TRIM24 (tripartite motif-containing protein 24) and BRPF1 (bromodomain and PHD finger containing protein 1) are epigenetics "readers" and potential therapeutic targets for cancer and other diseases. Here we describe the structure-guided design of 1-(indolin-1-yl)ethan-1-ones as novel TRIM24/BRPF1 bromodomain inhibitors. The representative compound 20l (Y08624) is a new TRIM24/BRPF1 dual inhibitor, with IC50 values of 0.98 and 1.16 µM, respectively. Cellular activity of 20l was validated by viability assay in prostate cancer (PC) cell lines. In PC xenograft models, 20l suppressed tumor growth (50 mg/kg/day, TGI = 53%) without exhibiting noticeable toxicity. Compound 20l represents a versatile starting point for the development of more potent TRIM24/BRPF1 inhibitors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte , Proteínas de Ligação a DNA , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Animais , Proteínas de Transporte/antagonistas & inibidores , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/antagonistas & inibidores , Humanos , Masculino , Neoplasias da Próstata/tratamento farmacológico , Domínios Proteicos
3.
Molecules ; 27(4)2022 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-35209054

RESUMO

The interaction between erlotinib (ERL) and bovine serum albumin (BSA) was studied in the presence of quercetin (QUR), a flavonoid with antioxidant properties. Ligands bind to the transport protein BSA resulting in competition between different ligands and displacing a bound ligand, resulting in higher plasma concentrations. Therefore, various spectroscopic experiments were conducted in addition to in silico studies to evaluate the interaction behavior of the BSA-ERL system in the presence and absence of QUR. The quenching curve and binding constants values suggest competition between QUR and ERL to bind to BSA. The binding constant for the BSA-ERL system decreased from 2.07 × 104 to 0.02 × 102 in the presence of QUR. The interaction of ERL with BSA at Site II is ruled out based on the site marker studies. The suggested Site on BSA for interaction with ERL is Site I. Stability of the BSA-ERL system was established with molecular dynamic simulation studies for both Site I and Site III interaction. In addition, the analysis can significantly help evaluate the effect of various quercetin-containing foods and supplements during the ERL-treatment regimen. In vitro binding evaluation provides a cheaper alternative approach to investigate ligand-protein interaction before clinical studies.


Assuntos
Proteínas de Transporte/química , Interações Medicamentosas , Cloridrato de Erlotinib/química , Cloridrato de Erlotinib/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Quercetina/química , Antioxidantes/química , Antioxidantes/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Quercetina/farmacologia , Análise Espectral , Relação Estrutura-Atividade
4.
J Am Chem Soc ; 144(2): 701-708, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-34994556

RESUMO

Proteolysis-targeting chimeras (PROTACs), heterobifunctional compounds that consist of protein-targeting ligands linked to an E3 ligase recruiter, have arisen as a powerful therapeutic modality for targeted protein degradation (TPD). Despite the popularity of TPD approaches in drug discovery, only a small number of E3 ligase recruiters are available for the >600 E3 ligases that exist in human cells. Here, we have discovered a cysteine-reactive covalent ligand, EN106, that targets FEM1B, an E3 ligase recently discovered as the critical component of the cellular response to reductive stress. By targeting C186 in FEM1B, EN106 disrupts recognition of the key reductive stress substrate of FEM1B, FNIP1. We further establish that EN106 can be used as a covalent recruiter for FEM1B in TPD applications by demonstrating that a PROTAC linking EN106 to the BET bromodomain inhibitor JQ1 or the kinase inhibitor dasatinib leads to the degradation of BRD4 and BCR-ABL, respectively. Our study showcases a covalent ligand that targets a natural E3 ligase-substrate binding site and highlights the utility of covalent ligand screening in expanding the arsenal of E3 ligase recruiters suitable for TPD applications.


Assuntos
Acetamidas/química , Proteínas de Ciclo Celular/metabolismo , Proteólise , Complexos Ubiquitina-Proteína Ligase/metabolismo , Animais , Azepinas/química , Sítios de Ligação , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular , Cisteína/química , Dasatinibe/química , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Inibidores de Proteínas Quinases/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Triazóis/química , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores , Complexos Ubiquitina-Proteína Ligase/genética
5.
Exp Neurol ; 348: 113944, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34896115

RESUMO

Fibroblast growth factor binding protein 3 (Fgfbp3) have been known to be crucial for the process of neural proliferation, differentiation, migration, and adhesion. However, the specific role and the molecular mechanisms of fgfbp3 in regulating the development of motor neurons remain unclear. In this study, we have investigated the function of fgfbp3 in morphogenesis and regeneration of motor neuron in zebrafish. Firstly, we found that fgfbp3 was localized in the motor neurons and loss of fgfbp3 caused the significant decrease of the length and branching number of the motor neuron axons, which could be partially rescued by fgfbp3 mRNA injection. Moreover, the fgfbp3 knockdown (KD) embryos demonstrated similar defects of motor neurons as identified in fgfbp3 knockout (KO) embryos. Furthermore, we revealed that the locomotion and startle response of fgfbp3 KO embryos were significantly restricted, which were partially rescued by the fgfbp3 overexpression. In addition, fgfbp3 KO remarkably compromised axonal regeneration of motor neurons after injury. Lastly, the malformation of motor neurons in fgfbp3 KO embryos was rescued by overexpressing drd1b or neurod6a, respectively, which were screened by transcriptome sequencing. Taken together, our results provide strong cellular and molecular evidence that fgfbp3 is crucial for the axonal morphogenesis and regeneration of motor neurons in zebrafish.


Assuntos
Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Neurônios Motores/metabolismo , Regeneração Nervosa/fisiologia , Neurogênese/fisiologia , Medula Espinal/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/antagonistas & inibidores , Técnicas de Inativação de Genes/métodos , Reflexo de Sobressalto/fisiologia , Medula Espinal/crescimento & desenvolvimento , Natação/fisiologia , Peixe-Zebra
6.
Drugs ; 82(1): 71-76, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34813049

RESUMO

Maralixibat (Livmarli™) is an orally-administered, small-molecule ileal bile acid transporter (IBAT) inhibitor being developed by Mirum Pharmaceuticals for the treatment of rare cholestatic liver diseases including Alagille syndrome (ALGS), progressive familial intrahepatic cholestasis (PFIC) and biliary atresia. Maralixibat received its first approval on 29 September 2021, in the USA, for use in the treatment of cholestatic pruritus in patients with ALGS 1 year of age and older. Maralixibat is also under regulatory review for ALGS in Europe, and clinical development for cholestatic liver disorders including ALGS in patients under 1 year of age, PFIC and biliary atresia is continuing in several other countries. This article summarises the milestones in the development of maralixibat leading to this first approval for ALGS.


Assuntos
Benzotiepinas , Proteínas de Transporte , Colestase Intra-Hepática , Glicoproteínas de Membrana , Humanos , Síndrome de Alagille/tratamento farmacológico , Atresia Biliar/tratamento farmacológico , Proteínas de Transporte/antagonistas & inibidores , Colestase Intra-Hepática/tratamento farmacológico , Ensaios Clínicos como Assunto , Aprovação de Drogas , Glicoproteínas de Membrana/antagonistas & inibidores , Estados Unidos , United States Food and Drug Administration , Benzotiepinas/administração & dosagem , Benzotiepinas/farmacologia , Benzotiepinas/uso terapêutico
7.
J Med Chem ; 65(2): 1171-1205, 2022 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-34726055

RESUMO

The allosteric regulation of pyruvate kinase M2 (PKM2) affects the switching of the PKM2 protein between the high-activity and low-activity states that allow ATP and lactate production, respectively. PKM2, in its low catalytic state (dimeric form), is chiefly active in metabolically energetic cells, including cancer cells. More recently, PKM2 has emerged as an attractive target due to its role in metabolic dysfunction and other interrelated conditions. PKM2 (dimer) activity can be inhibited by modulating PKM2 dimer-tetramer dynamics using either PKM2 inhibitors that bind at the ATP binding active site of PKM2 (dimer) or PKM2 activators that bind at the allosteric site of PKM2, thus activating PKM2 from the dimer formation to the tetrameric formation. The present perspective focuses on medicinal chemistry approaches to design and discover PKM2 inhibitors and activators and further provides a scope for the future design of compounds targeting PKM2 with better efficacy and selectivity.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Química Farmacêutica , Glicólise , Proteínas de Membrana/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Regulação Alostérica , Sítio Alostérico , Humanos , Hormônios Tireóideos
8.
Toxicol Appl Pharmacol ; 434: 115825, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34902352

RESUMO

Dyslipidemia or its severe version like familial hypercholesterolemia causes a high risk for cardiovascular diseases. Lomitapide, a microsomal triglyceride transfer protein inhibitor, is approved to treat familial hypercholesterolemia, associated with liver fat accumulation. In this work, we investigated the effect of the combination of lomitapide and triiodothyronine (T3) in Zucker fatty rats. Lomitapide (1 mg/kg, PO), or T3 (13 µg/kg, PO), or their combination, were given to these rats once daily for fourteen days. Body weight and food intake were recorded once daily during the treatment period. Serum and hepatic lipids, glucose tolerance, serum aminotransferases, bile fluids, hepatic gene expression, and liver histology were assessed at the end of the treatment. Lomitapide treatment reduced body weight, food intake, glucose intolerance, and serum lipids, and elevated serum aminotransferases and liver lipids. When combined with T3, lomitapide showed an enhanced reduction in body weight, food intake, serum cholesterol, serum LDL, and glucose intolerance. The combination treatment increased bile flow rate and biliary cholesterol excretion rate. Combining T3 with lomitapide attenuated the elevation of serum aminotransferases and liver lipids. Hepatic ABCB11, ABCG5, ABCG8, CYP7A1, CPT1, and ACOX1 expressions were increased with combination treatment. Histological analysis indicated that T3 attenuated hepatic fat accumulation caused by lomitapide. These data suggests that combining lomitapide with T3 may reduce lomitapide-induced hepatic toxicity and provide additional benefits in obesity and glucose intolerance.


Assuntos
Benzimidazóis/toxicidade , Ácidos e Sais Biliares/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Tri-Iodotironina/farmacologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Ratos , Ratos Zucker
9.
J Transl Med ; 19(1): 505, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34886858

RESUMO

Glioblastoma (GBM) is the most aggressive brain tumor of the central nervous system. Recent studies have reported the crucial functions of Tripartite Motif Containing 24 (TRIM24) in promoting cancer progression of GBM. However, it remains unclear if TRIM24 is an attractive druggable target for therapeutic intervention in GBM. We therefore performed a series of experiments, aiming to verify whether specific TRIM24 inhibition suppresses GBM malignant functions using dTRIM24 and IACS-9571, two novel selective TRIM24 antagonists. Our data showed that TRIM24 inhibitors serve as effective agents for inhibiting cell propagation and invasion of several patient-derived GBM stem cells (GSCs), and these effects are mediated partially through suppression of the TRIM24-SOX2 axis. This study provides novel insight into the TRIM24-based druggable dependencies, important for developing effective therapeutic strategies for brain tumors.


Assuntos
Neoplasias Encefálicas , Proteínas de Transporte/antagonistas & inibidores , Glioblastoma , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células Tumorais Cultivadas
10.
Microbiol Spectr ; 9(3): e0095121, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34908453

RESUMO

Staphylococcus aureus is a versatile human commensal bacteria and pathogen that causes various community and hospital-acquired infections. The S. aureus efflux pump NorA which belongs to the major facilitator superfamily, confers resistance to a range of substrates. Many efflux pump inhibitors (EPIs) have been discovered, but none is clinically approved due to their undesirable toxicities. In this study, we have screened clinically approved drugs for possible NorA EPI-like activity. We identified six drugs that showed the best efflux pump inhibition in vitro, with a fractional inhibitory concentration index of ≤0.5, indicating synergism with hydrophilic fluoroquinolones. The mechanistic validation of efflux inhibitory potential was demonstrated in ethidium bromide-based accumulation and efflux inhibition assays. We further confirmed the functionality of EPIs by norfloxacin accumulation assay depicting more realistic proof of the conjecture. None of the EPIs disturbed membrane function or depleted the ATP synthesis levels in bacteria. Both raloxifene and pyrvinium displayed an increase in bactericidal activity of ciprofloxacin in time-kill kinetics, prolonged its post-antibiotic effect, and reduced the frequency of spontaneous resistant mutant development. The combination of EPIs with ciprofloxacin caused significant eradication of preformed biofilms. Moreover, in the murine thigh infection model, a single dose of pyrvinium combined with ciprofloxacin reduced the bacterial burden significantly compared to untreated control and ciprofloxacin alone, indicating the efficacy of the combination. Conclusively, this study represents approved drugs that can be repurposed and combined with antibiotics as NorA EPIs, having anti-biofilm properties to treat severe S. aureus infections at clinically relevant concentrations. IMPORTANCE Staphylococcus aureus is a frequent pathogen bacterium and the predominant cause of worsened nosocomial infections. Efflux pumps contribute to drug efflux and are reportedly associated with biofilm formation, thereby promoting difficult-to-treat biofilm-associated S. aureus infections. One strategy to combat these bacteria is to reduce active efflux and increase pathogen sensitivity to existing antibiotics. Repurposing approved drugs may solve the classical toxicity issues with previous efflux pump inhibitors and help reach sufficient plasma concentrations. We describe the in silico-based screening of FDA-approved drugs that identified six different molecules able to inhibit NorA pump (Major Facilitator Superfamily). Our study highlights that these compounds bind to and block the activity of the NorA pump and increase the sensitivity of S. aureus and methicillin-resistant S. aureus to fluoroquinolones. These drugs combined with fluoroquinolones significantly reduced the preformed biofilms and displayed significant efficacy in the murine thigh infection model when compared to untreated control and ciprofloxacin alone.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Transporte/antagonistas & inibidores , Reposicionamento de Medicamentos , Fluoroquinolonas/farmacologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Ciprofloxacina/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Farmacorresistência Bacteriana Múltipla/genética , Quimioterapia Combinada , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Norfloxacino/farmacologia , Compostos de Pirvínio/farmacologia , Cloridrato de Raloxifeno/farmacologia , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento
11.
Front Immunol ; 12: 779787, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899740

RESUMO

Rheumatoid arthritis (RA) is a classic autoimmune disease characterized by uncontrolled synovial proliferation, pannus formation, cartilage injury, and bone destruction. The specific pathogenesis of RA, a chronic inflammatory disease, remains unclear. However, both key glycolysis rate-limiting enzymes, hexokinase-II (HK-II), phosphofructokinase-1 (PFK-1), and pyruvate kinase M2 (PKM2), as well as indirect rate-limiting enzymes, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), are thought to participate in the pathogenesis of RA. In here, we review the latest literature on the pathogenesis of RA, introduce the pathophysiological characteristics of HK-II, PFK-1/PFKFB3, and PKM2 and their expression characteristics in this autoimmune disease, and systematically assess the association between the glycolytic rate-limiting enzymes and RA from a molecular level. Moreover, we highlight HK-II, PFK-1/PFKFB3, and PKM2 as potential targets for the clinical treatment of RA. There is great potential to develop new anti-rheumatic therapies through safe inhibition or overexpression of glycolysis rate-limiting enzymes.


Assuntos
Artrite Reumatoide/enzimologia , Enzimas/metabolismo , Glucose/metabolismo , Glicólise , Articulações/enzimologia , Animais , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Inibidores Enzimáticos/uso terapêutico , Glicólise/efeitos dos fármacos , Hexoquinase/antagonistas & inibidores , Hexoquinase/metabolismo , Humanos , Articulações/efeitos dos fármacos , Articulações/imunologia , Cinética , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Fosfofrutoquinase-1/antagonistas & inibidores , Fosfofrutoquinase-1/metabolismo , Fosfofrutoquinase-2/antagonistas & inibidores , Fosfofrutoquinase-2/metabolismo , Hormônios Tireóideos/metabolismo
12.
Mediators Inflamm ; 2021: 1805147, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790063

RESUMO

Mycobacterium tuberculosis (Mtb) remains a significant threat to global health as it induces granuloma and systemic inflammatory responses during active tuberculosis. Mtb can induce macrophage pyroptosis, leading to the release of IL-1ß and tissue damage, promoting its spread. Here, we established an in vitro Mtb-infected macrophage model to seek an effective antipyroptosis agent. Baicalin, isolated from Radix Scutellariae, was found to reduce pyroptosis in Mtb-infected macrophages. Baicalin could inhibit activation of the PERK/eIF2α pathway and thus downregulates TXNIP expression and subsequently reduces activation of the NLRP3 inflammasome, resulting in reduced pyroptosis in Mtb-infected macrophages. In conclusion, baicalin reduced pyroptosis by inhibiting the PERK/TXNIP/NLRP3 axis and might thus be a new adjuvant host-directed therapy (HDT) drug.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Flavonoides/farmacologia , Mycobacterium tuberculosis/patogenicidade , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Piroptose/efeitos dos fármacos , Tiorredoxinas/antagonistas & inibidores , eIF-2 Quinase/antagonistas & inibidores , Animais , Proteínas de Transporte/fisiologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Macrófagos/microbiologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Células RAW 264.7 , Tiorredoxinas/fisiologia , eIF-2 Quinase/fisiologia
13.
Front Immunol ; 12: 748573, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34759927

RESUMO

The cytokine storm is a marker of severity of various diseases and increased mortality. The altered metabolic profile and energy generation of immune cells affects their activation, exacerbating the cytokine storm. Currently, the emerging field of immunometabolism has highlighted the importance of specific metabolic pathways in immune regulation. The glycolytic enzyme pyruvate kinase M2 (PKM2) is a key regulator of immunometabolism and bridges metabolic and inflammatory dysfunction. This enzyme changes its conformation thus walks in different fields including metabolism and inflammation and associates with various transcription factors. This review summarizes the vital role of PKM2 in mediating immunometabolic reprogramming and its role in inducing cytokine storm, with a focus on providing references for further understanding of its pathological functions and for proposing new targets for the treatment of related diseases.


Assuntos
Proteínas de Transporte/fisiologia , Síndrome da Liberação de Citocina/enzimologia , Proteínas de Membrana/fisiologia , Hormônios Tireóideos/fisiologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proteínas de Transporte/agonistas , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/química , Núcleo Celular/enzimologia , Citocinas/metabolismo , Células Dendríticas/enzimologia , Ativação Enzimática , Ferroptose , Glicólise , Humanos , Inflamassomos , Inflamação , Células Matadoras Naturais/enzimologia , Macrófagos/enzimologia , Proteínas de Membrana/agonistas , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/química , Estrutura Molecular , Oxirredução , Conformação Proteica , Piroptose , Hormônios Tireóideos/agonistas , Hormônios Tireóideos/química
14.
Lancet ; 398(10311): 1581-1592, 2021 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-34755627

RESUMO

BACKGROUND: Alagille syndrome is a rare genetic disease that often presents with severe cholestasis and pruritus. There are no approved drugs for management. Maralixibat, an apical, sodium-dependent, bile acid transport inhibitor, prevents enterohepatic bile acid recirculation. We evaluated the safety and efficacy of maralixibat for children with cholestasis in Alagille syndrome. METHODS: ICONIC was a placebo-controlled, randomised withdrawal period (RWD), phase 2b study with open-label extension in children (aged 1-18 years) with Alagille syndrome (NCT02160782). Eligible participants had more than three times the normal serum bile acid (sBA) levels and intractable pruritus. After 18 weeks of maralixibat 380 µg/kg once per day, participants were randomly assigned (1:1) to continue maralixibat or receive placebo for 4 weeks. Subsequently, all participants received open-label maralixibat until week 48. During the long-term extension (204 weeks reported), doses were increased up to 380 µg/kg twice per day. The primary endpoint was the mean sBA change during the RWD in participants with at least 50% sBA reduction by week 18. Cholestastic pruritus was assessed using observer-rated, patient-rated, and clinician-rated 0-4 scales. The safety population was defined as all participants who had received at least one dose of maralixibat. This trial was registered with ClinicalTrials.gov, NCT02160782, and is closed to recruitment. FINDINGS: Between Oct 28, 2014, and Aug 14, 2015, 31 participants (mean age 5·4 years [SD 4·25]) were enrolled and 28 analysed at week 48. Of the 29 participants who entered the randomised drug withdrawal period, ten (34%) were female and 19 (66%) were male. In the RWD, participants switched to placebo had significant increases in sBA (94 µmol/L, 95% CI 23 to 164) and pruritus (1·7 points, 95% CI 1·2 to 2·2), whereas participants who continued maralixibat maintained treatment effect. This study met the primary endpoint (least square mean difference -117 µmol/L, 95% CI -232 to -2). From baseline to week 48, sBA (-96 µmol/L, -162 to -31) and pruritus (-1·6 pts, -2·1 to -1·1) improved. In participants who continued to week 204 (n=15) all improvements were maintained. Maralixibat was generally safe and well tolerated throughout. The most frequent adverse events were gastrointestinal related. Most adverse events were self-limiting in nature and mild-to-moderate in severity. INTERPRETATION: In children with Alagille syndrome, maralixibat is, to our knowledge, the first agent to show durable and clinically meaningful improvements in cholestasis. Maralixibat might represent a new treatment paradigm for chronic cholestasis in Alagille syndrome. FUNDING: Mirum Pharmaceuticals.


Assuntos
Síndrome de Alagille/tratamento farmacológico , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/uso terapêutico , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/uso terapêutico , Prurido/tratamento farmacológico , Adolescente , Proteínas de Transporte/efeitos adversos , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Glicoproteínas de Membrana/efeitos adversos , Resultado do Tratamento
15.
Molecules ; 26(22)2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34834098

RESUMO

Antimicrobial resistance (AMR) is a complex threat to human health and, to date, it represents a hot topic in drug discovery. The use of non-antibiotic molecules to block resistance mechanisms is a powerful alternative to the identification of new antibiotics. Bacterial efflux pumps exert the early step of AMR development, allowing the bacteria to grow in presence of sub-inhibitory drug concentration and develop more specific resistance mechanisms. Thus, efflux pump inhibitors (EPIs) offer a great opportunity to fight AMR, potentially restoring antibiotic activity. Based on our experience in designing and synthesizing novel EPIs, herein, we retrieved information around quinoline and indole derivatives reported in literature on this topic. Thus, our aim was to collect all data around these promising classes of EPIs in order to delineate a comprehensive structure-activity relationship (SAR) around each core for different microbes. With this review article, we aim to help future research in the field in the discovery of new microbial EPIs with improved activity and a better safety profile.


Assuntos
Antibacterianos , Bactérias , Infecções Bacterianas/tratamento farmacológico , Proteínas de Bactérias , Proteínas de Transporte , Quinolinas , Antibacterianos/química , Antibacterianos/uso terapêutico , Bactérias/química , Bactérias/metabolismo , Infecções Bacterianas/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Humanos , Quinolinas/química , Quinolinas/uso terapêutico , Relação Estrutura-Atividade
16.
Mol Med Rep ; 24(6)2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34643245

RESUMO

The aim of the present study was to investigate the effect of hedgehog­interacting protein antisense RNA 1 (HHIP­AS1) on epithelial­mesenchymal transition (EMT) and cellular stemness of human lung cancer cells by regulating the microRNA (miR)­153­3p/PCDHGA9 axis. Reverse transcription­quantitative PCR was used to compare the expression of HHIP­AS1 in lung cancer and adjacent normal lung tissues. In addition, the correlation of HHIP­AS1 with E­cadherin, Vimentin, N­cadherin and Twist1 was analyzed. HHIP­AS1 overexpression vector was transfected into lung cancer A549 and NCI­H1299 cell lines. Cell Counting Kit­8 and Transwell and clonogenic assays were used to detect the proliferation, invasion and clonogenesis of the lung cancer cells, respectively. The associations among HHIP­AS1, miR­153­3p and PCDHGA9 were predicted by bioinformatics analysis and verified by a dual­luciferase reporter system. The results showed that the expression of HHIP­AS1 in lung cancer tissues was significantly lower than that in normal tissues (P<0.001). HHIP­AS1 was positively correlated with E­cadherin and negatively correlated with Vimentin, N­cadherin and Twist1. HHIP­AS1 overexpression inhibited the proliferation, invasion and clonal formation of the A549 and NCI­H1299 cells. The luciferase reporter system verified that HHIP­AS1 could adsorb miR­153­3p and that PCDHGA9 was the target gene of miR­153­3p. A549 cells were transfected with HHIP­AS1 overexpression vector and miR­153­3p mimic, and the miR­153­3p mimic had a mitigating effect on HHIP­AS1 inhibition (P<0.001). In conclusion, HHIP­AS1 inhibits the EMT and stemness of lung cancer cells by regulating the miR­153­3p/PCDHGA9 axis. Thus, HHIP­AS1 may be a new potential target for lung cancer treatment.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Transição Epitelial-Mesenquimal/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Protocaderinas/genética , Protocaderinas/metabolismo , Células A549 , Idoso , Idoso de 80 Anos ou mais , Antígenos CD , Caderinas/genética , Caderinas/metabolismo , Proteínas de Transporte/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Antissenso , RNA Longo não Codificante/genética , Proteína 1 Relacionada a Twist , Vimentina
17.
Drug Metab Dispos ; 49(12): 1109-1117, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34625435

RESUMO

Linerixibat, an oral small-molecule ileal bile acid transporter inhibitor under development for cholestatic pruritus in primary biliary cholangitis, was designed for minimal absorption from the intestine (site of pharmacological action). This study characterized the pharmacokinetics, absorption, metabolism, and excretion of [14C]-linerixibat in humans after an intravenous microtracer concomitant with unlabeled oral tablets and [14C]-linerixibat oral solution. Linerixibat exhibited absorption-limited flip-flop kinetics: longer oral versus intravenous half-life (6-7 hours vs. 0.8 hours). The short intravenous half-life was consistent with high systemic clearance (61.9 l/h) and low volume of distribution (16.3 l). In vitro studies predicted rapid hepatic clearance via cytochrome P450 3A4 metabolism, which predicted human hepatic clearance within 1.5-fold. However, linerixibat was minimally metabolized in humans after intravenous administration: ∼80% elimination via biliary/fecal excretion (>90%-97% as unchanged parent) and ∼20% renal elimination by glomerular filtration (>97% as unchanged parent). Absolute oral bioavailability of linerixibat was exceedingly low (0.05%), primarily because of a very low fraction absorbed (0.167%; fraction escaping first-pass gut metabolism (fg) ∼100%), with high hepatic extraction ratio (77.0%) acting as a secondary barrier to systemic exposure. Oral linerixibat was almost entirely excreted (>99% recovered radioactivity) in feces as unchanged and unabsorbed linerixibat. Consistent with the low oral fraction absorbed and ∼20% renal recovery of intravenous [14C]-linerixibat, urinary elimination of orally administered radioactivity was negligible (<0.04% of dose). Linerixibat unequivocally exhibited minimal gastrointestinal absorption and oral systemic exposure. Linerixibat represents a unique example of high CYP3A4 clearance in vitro but nearly complete excretion as unchanged parent drug via the biliary/fecal route. SIGNIFICANCE STATEMENT: This study conclusively established minimal absorption and systemic exposure to orally administered linerixibat in humans. The small amount of linerixibat absorbed was eliminated efficiently as unchanged parent drug via the biliary/fecal route. The hepatic clearance mechanism was mispredicted to be mediated via cytochrome P450 3A4 metabolism in vitro rather than biliary excretion of unchanged linerixibat in vivo.


Assuntos
Administração Intravenosa , Administração Oral , Proteínas de Transporte/antagonistas & inibidores , Eliminação Hepatobiliar , Glicoproteínas de Membrana/antagonistas & inibidores , Metilaminas/farmacocinética , Eliminação Renal , Tiazepinas/farmacocinética , Adulto , Disponibilidade Biológica , Fármacos Gastrointestinais/farmacocinética , Voluntários Saudáveis , Eliminação Hepatobiliar/efeitos dos fármacos , Eliminação Hepatobiliar/fisiologia , Humanos , Absorção Intestinal , Masculino , Taxa de Depuração Metabólica , Eliminação Renal/efeitos dos fármacos , Eliminação Renal/fisiologia , Resultado do Tratamento
18.
Angew Chem Int Ed Engl ; 60(51): 26755-26761, 2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34626154

RESUMO

Cholesterol transport proteins regulate a vast array of cellular processes including lipid metabolism, vesicular and non-vesicular trafficking, organelle contact sites, and autophagy. Despite their undoubted importance, the identification of selective modulators of this class of proteins has been challenging due to the structural similarities in the cholesterol-binding site. Herein we report a general strategy for the identification of selective inhibitors of cholesterol transport proteins via the synthesis of a diverse sterol-inspired compound collection. Fusion of a primary sterol fragment to an array of secondary privileged scaffolds led to the identification of potent and selective inhibitors of the cholesterol transport protein Aster-C, which displayed a surprising preference for the unnatural-sterol AB-ring stereochemistry and new inhibitors of Aster-A. We propose that this strategy can and should be applied to any therapeutically relevant sterol-binding protein.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Colesterol/metabolismo , Esteróis/farmacologia , Transporte Biológico/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Humanos , Estrutura Molecular , Esteróis/síntese química , Esteróis/química
19.
Oxid Med Cell Longev ; 2021: 7397516, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603601

RESUMO

The death of nucleus pulposus (NP) cells is an important cause of intervertebral disc (IVD) degeneration. Redox disturbance caused by dysfunctional mitochondria has been considered as a vital risk for NP cell survival. It is valuable to identify key proteins maintaining mitochondrial function in NP cells. A previous study found that regulated in development and DNA damage response 1 (REDD1) are upregulated during intervertebral disc degeneration and that REDD1 can cause NP cell apoptosis. Thus, the present study further explores the effect of REDD1 on IVD degeneration. Our results showed that REDD1 promotes NP cell apoptosis via the mitochondrial pathway. Importantly, REDD1 formed a complex with TXNIP to strengthen its own action, and the combination was consolidated under H2O2-induced oxidative stress. The combined inhibition of the REDD1/TXNIP complex was better than that of REDD1 or TXNIP alone in restoring cell proliferation and accelerating apoptosis. Moreover, p53 acts as the transcription factor of REDD1 to regulate the REDD1/TXNIP complex under oxidative stress. Altogether, our results demonstrated that the REDD1/TXNIP complex mediated H2O2-induced human NP cell apoptosis and IVD degeneration through the mitochondrial pathway. Interferences on these sites to achieve mitochondrial redox homeostasis may be a novel therapeutic strategy for oxidative stress-associated IVD degeneration.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Fatores de Transcrição/metabolismo , Adolescente , Adulto , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Peróxido de Hidrogênio/farmacologia , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/patologia , Masculino , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Núcleo Pulposo/citologia , Núcleo Pulposo/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/metabolismo , Adulto Jovem
20.
Drugs ; 81(15): 1781-1786, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34499340

RESUMO

Odevixibat (Bylvay™) is a small molecule inhibitor of the ileal bile acid transporter being developed by Albireo Pharma, Inc. for the treatment of various cholestatic diseases, including progressive familial intrahepatic cholestasis (PFIC). In July 2021, odevixibat received its first approval in the EU for the treatment of PFIC in patients aged ≥ 6 months, followed shortly by its approval in the USA for the treatment of pruritus in patients aged ≥ 3 months with PFIC. Odevixibat is also in clinical development for the treatment of other cholestatic diseases, including Alagille syndrome and biliary atresia, in various countries. This article summarizes the milestones in the development of odevixibat leading to this first approval for PFIC.


Assuntos
Benzodiazepinas/uso terapêutico , Butiratos/uso terapêutico , Colestase/tratamento farmacológico , Prurido/tratamento farmacológico , Benzodiazepinas/farmacologia , Butiratos/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Colestase/fisiopatologia , Aprovação de Drogas , Desenvolvimento de Medicamentos , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Prurido/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...