Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
1.
JCI Insight ; 9(15)2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38900587

RESUMEN

Pathological deposition and crosslinking of collagen type I by activated myofibroblasts drives progressive tissue fibrosis. Therapies that inhibit collagen synthesis have potential as antifibrotic agents. We identify the collagen chaperone cyclophilin B as a major cellular target of the natural product sanglifehrin A (SfA) using photoaffinity labeling and chemical proteomics. Mechanistically, SfA inhibits and induces the secretion of cyclophilin B from the endoplasmic reticulum (ER) and prevents TGF-ß1-activated myofibroblasts from synthesizing and secreting collagen type I in vitro, without inducing ER stress or affecting collagen type I mRNA transcription, myofibroblast migration, contractility, or TGF-ß1 signaling. In vivo, SfA induced cyclophilin B secretion in preclinical models of fibrosis, thereby inhibiting collagen synthesis from fibrotic fibroblasts and mitigating the development of lung and skin fibrosis in mice. Ex vivo, SfA induces cyclophilin B secretion and inhibits collagen type I secretion from fibrotic human lung fibroblasts and samples from patients with idiopathic pulmonary fibrosis (IPF). Taken together, we provide chemical, molecular, functional, and translational evidence for demonstrating direct antifibrotic activities of SfA in preclinical and human ex vivo fibrotic models. Our results identify the cellular target of SfA, the collagen chaperone cyclophilin B, as a mechanistic target for the treatment of organ fibrosis.


Asunto(s)
Ciclofilinas , Animales , Humanos , Ratones , Ciclofilinas/metabolismo , Ciclofilinas/antagonistas & inhibidores , Colágeno Tipo I/metabolismo , Fibrosis , Miofibroblastos/metabolismo , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta1/metabolismo , Lactonas , Compuestos de Espiro
2.
Brain ; 147(5): 1710-1725, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38146639

RESUMEN

Mitochondrial dysfunction is an early pathological feature of Alzheimer disease and plays a crucial role in the development and progression of Alzheimer's disease. Strategies to rescue mitochondrial function and cognition remain to be explored. Cyclophilin D (CypD), the peptidylprolyl isomerase F (PPIase), is a key component in opening the mitochondrial membrane permeability transition pore, leading to mitochondrial dysfunction and cell death. Blocking membrane permeability transition pore opening by inhibiting CypD activity is a promising therapeutic approach for Alzheimer's disease. However, there is currently no effective CypD inhibitor for Alzheimer's disease, with previous candidates demonstrating high toxicity, poor ability to cross the blood-brain barrier, compromised biocompatibility and low selectivity. Here, we report a new class of non-toxic and biocompatible CypD inhibitor, ebselen, using a conventional PPIase assay to screen a library of ∼2000 FDA-approved drugs with crystallographic analysis of the CypD-ebselen crystal structure (PDB code: 8EJX). More importantly, we assessed the effects of genetic and pharmacological blockade of CypD on Alzheimer's disease mitochondrial and glycolytic bioenergetics in Alzheimer's disease-derived mitochondrial cybrid cells, an ex vivo human sporadic Alzheimer's disease mitochondrial model, and on synaptic function, inflammatory response and learning and memory in Alzheimer's disease mouse models. Inhibition of CypD by ebselen protects against sporadic Alzheimer's disease- and amyloid-ß-induced mitochondrial and glycolytic perturbation, synaptic and cognitive dysfunction, together with suppressing neuroinflammation in the brain of Alzheimer's disease mouse models, which is linked to CypD-related membrane permeability transition pore formation. Thus, CypD inhibitors have the potential to slow the progression of neurodegenerative diseases, including Alzheimer's disease, by boosting mitochondrial bioenergetics and improving synaptic and cognitive function.


Asunto(s)
Enfermedad de Alzheimer , Isoindoles , Mitocondrias , Compuestos de Organoselenio , Peptidil-Prolil Isomerasa F , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Peptidil-Prolil Isomerasa F/metabolismo , Animales , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratones , Humanos , Cognición/efectos de los fármacos , Azoles/farmacología , Azoles/uso terapéutico , Ciclofilinas/metabolismo , Ciclofilinas/antagonistas & inhibidores , Ratones Transgénicos , Ratones Endogámicos C57BL , Masculino , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico
3.
PLoS One ; 16(11): e0259374, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34843507

RESUMEN

Whitefly, Bemisia tabaci (Gennadius) is an important pest of cotton causing direct damage as sap feeder and vector of Cotton leaf curl virus (CLCuV). Previous few studies suggest that female whiteflies are more efficient vector of begomovirusthan males, however the sex-biased transmission efficiency is still not clearly understood. Present studies with B. tabaci AsiaII-1 haplotype showed higher virus transmission efficiency of females compared to males. This variable begomovirus transmission efficiency has been related to previously identifiedkey factors associated with B. tabaci. The higher density of endosymbiont Arsenophonus and variable expression of some midgut proteins genes i.e. Cyclophilin, Knottin, Hsp40, Hsp70 may be possibly imparting higher vector competency to the females compared to males. The present studies suggest low abundance of Arsenophonus spp. as well as lower expressionof Cyclophilin genein males as compared to females. This is further supplemented by overexpression of Knottin, Hsp40, and Hsp70 genes in males compared to females and thus collectively all these factors might be playing a key role in low virus transmission efficiency of males. The relative density of Arsenophonus spp. and expression of midgut proteins genes in male and female whitefly first time enriches our understanding about sex-biased transmission efficiency of begomovirus.


Asunto(s)
Begomovirus/fisiología , Sistema Digestivo/metabolismo , Gammaproteobacteria/crecimiento & desarrollo , Hemípteros/virología , Proteínas de Insectos/metabolismo , Animales , Begomovirus/crecimiento & desarrollo , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/genética , Ciclofilinas/metabolismo , Femenino , Gammaproteobacteria/aislamiento & purificación , Gammaproteobacteria/fisiología , Regulación de la Expresión Génica , Silenciador del Gen , Proteínas del Choque Térmico HSP40/antagonistas & inhibidores , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Haplotipos , Proteínas de Insectos/antagonistas & inhibidores , Proteínas de Insectos/genética , Insectos Vectores/virología , Masculino , Enfermedades de las Plantas/virología , ARN Bicatenario/metabolismo , Factores Sexuales , Simbiosis , Carga Viral
4.
PLoS One ; 16(5): e0251934, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34014993

RESUMEN

We and others previously reported that the direct-acting agents (DAA) NS5A inhibitors (NS5Ai) and the host-targeting agents cyclophilin inhibitors (CypIs) inhibit HCV replication in vitro. In this study, we investigated whether the combination of NS5Ai and CypI offers a potent anti-HCV effect in vivo. A single administration of NS5Ai or CypI alone to HCV-infected humanized-mice inhibits HCV replication. The combination of NS5Ai with CypI suppresses HCV (GT1a, GT2a, GT3a and GT4a) replication in an additive manner. NS5Ai/CypI combinations provide a statistically more profound anti-HCV inhibition for GT2a and GT3a than GT1a and GT4a, leading to a fastest and deepest inhibition of GT2a and GT3a replications. Combining CypI with NS5Ai prevents the viral rebound normally observed in mice treated with NS5Ai alone. Results were confirmed in mice implanted with human hepatocytes from different donors. Therefore, the combination of NS5Ai with CypI may serve as a regimen for the treatment of HCV patients with specific genotypes and disorder conditions, which diminish sustain viral response levels to DAA, such as GT3a infection, cirrhosis, and DAA resistance associated with the selection of resistance-associated substitutions present at baseline or are acquired during treatment.


Asunto(s)
Antivirales/farmacología , Ciclofilinas/genética , Hepacivirus/efectos de los fármacos , Cirrosis Hepática/tratamiento farmacológico , Animales , Ciclofilinas/antagonistas & inhibidores , Farmacorresistencia Viral/genética , Genotipo , Hepacivirus/patogenicidad , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/virología , Ratones , Proteínas no Estructurales Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
5.
Naunyn Schmiedebergs Arch Pharmacol ; 394(5): 941-954, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33284399

RESUMEN

Binary enterotoxins Clostridioides difficile CDT toxin, Clostridium botulinum C2 toxin, and Clostridium perfringens iota toxin consist of two separate protein components. The B-components facilitate receptor-mediated uptake into mammalian cells and form pores into endosomal membranes through which the enzymatic active A-components translocate into the cytosol. Here, the A-components ADP-ribosylate G-actin which leads to F-actin depolymerization followed by rounding of cells which causes clinical symptoms. The protein folding helper enzymes Hsp90, Hsp70, and peptidyl-prolyl cis/trans isomerases of the cyclophilin (Cyp) and FK506 binding protein (FKBP) families are required for translocation of A-components of CDT, C2, and iota toxins from endosomes to the cytosol. Here, we demonstrated that simultaneous inhibition of these folding helpers by specific pharmacological inhibitors protects mammalian, including human, cells from intoxication with CDT, C2, and iota toxins, and that the inhibitor combination displayed an enhanced effect compared to application of the individual inhibitors. Moreover, combination of inhibitors allowed a concentration reduction of the individual compounds as well as decreasing of the incubation time with inhibitors to achieve a protective effect. These results potentially have implications for possible future therapeutic applications to relieve clinical symptoms caused by bacterial toxins that depend on Hsp90, Hsp70, Cyps, and FKBPs for their membrane translocation into the cytosol of target cells.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Toxinas Bacterianas/toxicidad , Toxinas Botulínicas/toxicidad , Enterotoxinas/toxicidad , Animales , Células CACO-2 , Chlorocebus aethiops , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/metabolismo , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/metabolismo , Células Vero
6.
Eur Respir J ; 56(5)2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32616594

RESUMEN

While severe coronavirus infections, including Middle East respiratory syndrome coronavirus (MERS-CoV), cause lung injury with high mortality rates, protective treatment strategies are not approved for clinical use.We elucidated the molecular mechanisms by which the cyclophilin inhibitors cyclosporin A (CsA) and alisporivir (ALV) restrict MERS-CoV to validate their suitability as readily available therapy in MERS-CoV infection.Calu-3 cells and primary human alveolar epithelial cells (hAECs) were infected with MERS-CoV and treated with CsA or ALV or inhibitors targeting cyclophilin inhibitor-regulated molecules including calcineurin, nuclear factor of activated T-cells (NFATs) or mitogen-activated protein kinases. Novel CsA-induced pathways were identified by RNA sequencing and manipulated by gene knockdown or neutralising antibodies. Viral replication was quantified by quantitative real-time PCR and 50% tissue culture infective dose. Data were validated in a murine MERS-CoV infection model.Both CsA and ALV reduced MERS-CoV titres and viral RNA replication in Calu-3 cells and hAECs, improving epithelial integrity. While neither calcineurin nor NFAT inhibition reduced MERS-CoV propagation, blockade of c-Jun N-terminal kinase diminished infectious viral particle release but not RNA accumulation. Importantly, CsA induced interferon regulatory factor 1 (IRF1), a pronounced type III interferon (IFNλ) response and expression of antiviral genes. Downregulation of IRF1 or IFNλ increased MERS-CoV propagation in the presence of CsA. Importantly, oral application of CsA reduced MERS-CoV replication in vivo, correlating with elevated lung IFNλ levels and improved outcome.We provide evidence that cyclophilin inhibitors efficiently decrease MERS-CoV replication in vitro and in vivo via upregulation of inflammatory antiviral cell responses, in particular IFNλ. CsA might therefore represent a promising candidate for treating MERS-CoV infection.


Asunto(s)
Infecciones por Coronavirus/prevención & control , Ciclofilinas/antagonistas & inhibidores , Ciclosporina/farmacología , Interferones/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Animales , Inhibidores de la Calcineurina/farmacología , Técnicas de Cultivo de Célula , Infecciones por Coronavirus/metabolismo , Modelos Animales de Enfermedad , Humanos , Factor 1 Regulador del Interferón/efectos de los fármacos , Factor 1 Regulador del Interferón/metabolismo , Interferones/efectos de los fármacos , Ratones , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Replicación Viral/efectos de los fármacos , Interferón lambda
7.
Int J Med Sci ; 17(12): 1803-1810, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32714083

RESUMEN

Since the end of 2019, a new type of coronavirus pneumonia (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has been spreading rapidly throughout the world. Previously, there were two outbreaks of severe coronavirus caused by different coronaviruses worldwide, namely Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV). This article introduced the origin, virological characteristics and epidemiological overview of SARS-CoV-2, reviewed the currently known drugs that may prevent and treat coronavirus, explained the characteristics of the new coronavirus and provided novel information for the prevention and treatment of COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Amidas/farmacología , Amidas/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Betacoronavirus/aislamiento & purificación , Betacoronavirus/fisiología , COVID-19 , Cloroquina/análogos & derivados , Cloroquina/uso terapéutico , Clorpromazina/uso terapéutico , Coronavirus/genética , Infecciones por Coronavirus/genética , Ciclofilinas/antagonistas & inhibidores , Desarrollo de Medicamentos , Reposicionamiento de Medicamentos , Medicamentos Herbarios Chinos/uso terapéutico , Endocitosis/efectos de los fármacos , Humanos , Sueros Inmunes , Inductores de Interferón/uso terapéutico , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Neumonía Viral/genética , Pirazinas/farmacología , Pirazinas/uso terapéutico , Resveratrol/farmacología , Resveratrol/uso terapéutico , SARS-CoV-2 , Vacunas Virales/uso terapéutico , Tratamiento Farmacológico de COVID-19
8.
Acc Chem Res ; 53(9): 1782-1790, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32658452

RESUMEN

RNA-based technologies to control gene expression, such as RNA interference (RNAi) and CRISPR-Cas9, have become powerful tools in molecular biology and genomics. The exciting potential that RNAi and CRISPR-Cas9 may also become new therapeutic approaches has reinvigorated interest in chemically modifying RNA to improve its properties for in vivo applications. Chemical modifications can improve enzymatic stability, in vivo delivery, cellular uptake, and sequence specificity as well as minimize off-target activity of short interfering RNAs (siRNAs) and CRISPR associated RNAs. While numerous good solutions for improving stability toward enzymatic degradation have emerged, optimization of the latter functional properties remains challenging. In this Account, we discuss synthesis, structure, and biological activity of novel nonionic analogues of RNA that have the phosphodiester backbone replaced by amide linkages (AM1). Our long-term goal is to use the amide backbone to improve the stability and specificity of siRNAs and other functional RNAs. Our work in this area was motivated by early discoveries that nonionic backbone modifications, including AM1, did not disturb the overall structure or thermal stability of RNA duplexes. We hypothesized that the reduced negative charge and hydrophobic nature of the AM1 backbone modification might be useful in optimizing functional applications through enhanced cellular uptake, and might suppress unwanted off-target effects of siRNAs. NMR and X-ray crystallography studies showed that AM1 was an excellent mimic of phosphodiester linkages in RNA. The local conformational changes caused by the amide linkages were easily accommodated by small adjustments in RNA's conformation. Further, the amide carbonyl group assumed an orientation that is similar to one of the nonbridging P-O bonds, which may enable amide/phosphate mimicry by conserving hydrogen bonding interactions. The crystal structure of a short amide-modified DNA-RNA hybrid in complex with RNase H indicated that the amide N-H could also act as an H-bond donor to stabilize RNA-protein interactions, which is an interaction mode not available to phosphate groups. Functional assays established that amides were well tolerated at internal positions in both strands of siRNAs. Surprisingly, amide modifications in the middle of the guide strand and at the 5'-end of the passenger strand increased RNAi activity compared to unmodified siRNA. Most importantly, an amide linkage between the first and second nucleosides of the passenger strand completely abolished its undesired off-target activity while enhancing the desired RNAi activity. These results suggest that RNAi may tolerate more substantial modifications of siRNAs than the chemistries tried so far. The findings are also important and timely because they demonstrate that amide modifications may reduce off-target activity of siRNAs, which remains an important roadblock for clinical use of RNAi. Taken together, our work suggests that amide linkages have underappreciated potential to optimize the biological and pharmacological properties of RNA. Expanded use of amide linkages in RNA to enhance CRISPR and other technologies requiring chemically stable, functional mimics of noncoding RNAs is expected.


Asunto(s)
Amidas/química , Interferencia de ARN , ARN Interferente Pequeño/química , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/genética , Ciclofilinas/metabolismo , Dimerización , Edición Génica , Células HeLa , Humanos , Nucleósidos/química , Estabilidad del ARN , ARN Interferente Pequeño/metabolismo
9.
Bioorg Med Chem Lett ; 30(18): 127423, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32731088

RESUMEN

Evidence that hepatitis C virus (HCV) utilizes cellular cyclophilin proteins in the virus replication cycle has increased attention on cyclophilin inhibitors as attractive therapeutic targets in the treatment of HCV. Previous reports have described a number of non-immunosuppressive cyclophilin inhibitors, most of which require many synthetic steps for their preparation. Sasamura et al. have previously reported the isolation of bioconversion derivative 4. This analog is a convenient starting point for optimization due to the presence of the readily modifiable primary hydroxyl group and because it shows moderate anti-HCV activity and decreased immunosuppressive activity. We have also established an efficient C-alkylation reaction at the 3-position. Through a detailed structure-activity relationship study, we discovered a new type of clinical candidate 14 which requires a short synthetic process and has potent anti-HCV activity and reduced immunosuppressive activity, as well as improved aqueous solubility and pharmacokinetics.


Asunto(s)
Antivirales/síntesis química , Ciclofilinas/antagonistas & inhibidores , Ciclosporina/síntesis química , Inhibidores Enzimáticos/síntesis química , Hepatitis C/tratamiento farmacológico , Alquilación , Secuencia de Aminoácidos , Antivirales/farmacocinética , Ciclosporina/química , Ciclosporina/farmacocinética , Ciclosporina/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Humanos , Inmunosupresores/farmacología , Solubilidad , Relación Estructura-Actividad
10.
Bioorg Med Chem Lett ; 30(16): 127308, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32631528

RESUMEN

Evidence indicates that hepatitis C virus (HCV) utilizes cellular cyclophilin proteins in its replication, and cyclophilin inhibitors represent a new class of anti-HCV agents. We have established an efficient synthetic methodology to generate FR901459 derivatives via N, O-acyl migration reaction while avoiding total synthesis. Through a detailed structure-activity relationship study, we improved anti-HCV activity while decreasing immunosuppressive activity. Additionally, we discovered the importance of substitution at the 3 position for not only improving anti-HCV activity but also pharmacokinetic profile. Finally, by striking an appropriate balance between potency, solubility, and permeability, we discovered ASP5286 (13) as a potential clinical candidate for anti-HCV therapy.


Asunto(s)
Antivirales/farmacología , Ciclofilinas/antagonistas & inhibidores , Descubrimiento de Drogas , Hepacivirus/efectos de los fármacos , Inmunosupresores/farmacología , Antivirales/síntesis química , Antivirales/química , Relación Dosis-Respuesta a Droga , Inmunosupresores/síntesis química , Inmunosupresores/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad
11.
Bioorg Med Chem Lett ; 30(14): 127251, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32527551

RESUMEN

HCV utilizes cellular protein cyclophilins in the virus replication cycle and cyclophilin inhibitors have become a new class of anti-HCV agents. In our screening of natural products, we identified a unique cyclosporin analogue, FR901459, as a cyclophilin inhibitor with potent anti-HCV activity. In this work, we developed an efficient synthetic methodology to prepare FR901459 derivatives via an N, O-acyl migration reaction. This method allows us to efficiently manipulate the amino acid residues at the 3 position while avoiding lengthy total synthesis for each compound. By using this methodology, we discovered 4, which has superior anti-HCV activity and decreased immunosuppressive activity compared to FR901459.


Asunto(s)
Antivirales/farmacología , Ciclosporina/farmacología , Desarrollo de Medicamentos , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Inmunosupresores/farmacología , Antivirales/síntesis química , Antivirales/química , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/metabolismo , Ciclosporina/síntesis química , Ciclosporina/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Inmunosupresores/síntesis química , Inmunosupresores/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad
12.
Artículo en Inglés | MEDLINE | ID: mdl-32376613

RESUMEN

Cyclophilins play a key role in the life cycle of coronaviruses. Alisporivir (Debio 025) is a nonimmunosuppressive analogue of cyclosporine with potent cyclophilin inhibition properties. Alisporivir reduced SARS-CoV-2 RNA production in a dose-dependent manner in Vero E6 cells, with a 50% effective concentration (EC50) of 0.46 ± 0.04 µM. Alisporivir inhibited a postentry step of the SARS-CoV-2 life cycle. These results justify rapidly conducting a proof-of-concept phase 2 trial with alisporivir in patients with SARS-CoV-2 infection.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Ciclofilinas/antagonistas & inhibidores , Ciclosporina/farmacología , Neumonía Viral/tratamiento farmacológico , Animales , Antivirales/farmacología , COVID-19 , Línea Celular , Chlorocebus aethiops , Humanos , Pandemias , SARS-CoV-2 , Células Vero , Replicación Viral/efectos de los fármacos
13.
Clin Infect Dis ; 71(16): 2191-2194, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32409832

RESUMEN

December 2019 saw the emergence of a new epidemic of pneumonia of varying severity, called coronavirus disease 2019 (COVID-19), caused by a newly identified coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV-2). No therapeutic option is available to treat this infection that has already killed > 310 000 people worldwide. This Viewpoint summarizes the strong scientific arguments supporting the use of alisporivir, a nonimmunosuppressive analogue of cyclosporine A with potent cyclophilin inhibition properties that has reached phase 3 clinical development, for the treatment of COVID-19. They include the strong cyclophilin dependency of the life cycle of many coronaviruses, including severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome coronavirus, and preclinical data showing strong antiviral and cytoprotective properties of alisporivir in various models of coronavirus infection, including SARS-CoV-2. Alisporivir should be tested without delay on both virological and clinical endpoints in patients with or at risk of severe forms of SARS-CoV-2 infection.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Ciclofilinas/antagonistas & inhibidores , Ciclosporina/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Animales , COVID-19/epidemiología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Ratones , Ratas
14.
Neuropharmacology ; 171: 108088, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32277944

RESUMEN

The interaction between apoptosis-inducing factor (AIF) and cyclophilin A (CypA) has been shown to contribute to caspase-independent apoptosis. Blocking the AIF/CypA interaction protects against glutamate-induced neuronal cell death in vitro, and the purpose of this study was to determine the in vivo effect of an AIF/CypA interaction blocking peptide (AIF(370-394)-TAT) on neonatal mouse brain injury after hypoxia-ischemia (HI). The pups were treated with AIF (370-394)-TAT peptide intranasally prior to HI. Brain injury was significantly reduced at 72 h after HI in the AIF(370-394)-TAT peptide treatment group compared to vehicle-only treatment for both the gray matter and the subcortical white matter, and the neuroprotection was more pronounced in males than in females. Neuronal cell death was evaluated in males at 8 h and 24 h post-HI, and it was decreased significantly in the CA1 region of the hippocampus and the nucleus habenularis region after AIF(370-394)-TAT treatment. Caspase-independent apoptosis was decreased in the cortex, striatum, and nucleus habenularis after AIF(370-394)-TAT treatment, but no significant change was found on caspase-dependent apoptosis as indicated by the number of active caspase-3-labeled cells. Further analysis showed that both AIF and CypA nuclear accumulation were decreased after treatment with the AIF(370-394)-TAT peptide. These results suggest that AIF(370-394)-TAT inhibited AIF/CypA translocation to the nucleus and reduced HI-induced caspase-independent apoptosis and brain injury in young male mice, suggesting that blocking AIF/CypA might be a potential therapeutic target for neonatal brain injury.


Asunto(s)
Factor Inductor de la Apoptosis/antagonistas & inhibidores , Factor Inductor de la Apoptosis/farmacología , Ciclofilinas/antagonistas & inhibidores , Hipoxia-Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/farmacología , Administración Intranasal , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Caspasas/fisiología , Muerte Celular/efectos de los fármacos , Femenino , Sustancia Gris/patología , Hipoxia-Isquemia Encefálica/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Biogénesis de Organelos , Caracteres Sexuales , Sustancia Blanca/patología
15.
FEBS J ; 287(20): 4500-4524, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32056376

RESUMEN

Evidence is emerging that inorganic polyphosphate (polyP) is a fundamental molecule involved in a wide range of biological processes. In higher eukaryotes, polyP is abundant in osteoblasts but questions remain as to its functions. Here, we find that polyP is particularly enriched in endoplasmic reticulum (ER) where it colocalizes with cyclophilin B (CypB) using osteoblastic SaOS-2 model cell line. PolyP binds directly and specifically to CypB, inhibiting its peptidyl-prolyl cis-trans isomerase activity which is critical for collagen folding. PolyP sequestration by spermine and ER-specific polyP reduction by polyphosphatase expression in cells reduced collagen misfolding and confirmed that endogenous polyP acts as a molecular control of CypB-mediated collagen folding. We propose that polyP is a previously unrecognized critical regulator of protein homeostasis in ER.


Asunto(s)
Colágeno/efectos de los fármacos , Ciclofilinas/antagonistas & inhibidores , Osteoblastos/efectos de los fármacos , Polifosfatos/farmacología , Pliegue de Proteína/efectos de los fármacos , Colágeno/metabolismo , Ciclofilinas/farmacología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Humanos , Osteoblastos/metabolismo , Células Tumorales Cultivadas
16.
Expert Opin Investig Drugs ; 29(2): 163-178, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31868526

RESUMEN

Introduction: Cyclophilins are a family of diverse regulatory enzymes that have been studied for over 30 years; they participate in many pathophysiological processes. Genetic deletion or pharmacologic inhibition of cyclophilins has shown therapeutic effects in a wide spectrum of disease models, including liver disorders, and hence may be beneficial in treating nonalcoholic steatohepatitis (NASH).Areas Covered: This articles briefly describes cyclophilin isomerases and the main classes of cyclophilin antagonists; it then summarizes data showing cyclophilin participation in the major pathophysiological activities that occur in NASH.Expert Opinion: Optimization of therapeutic outcomes in the treatment of NASH may be best realized by targeting multiple pathologic pathways, especially when treating advanced stages of the disease. A preferred approach for achieving this goal is to use compounds such as cyclophilin inhibitors that simultaneously target multiple disease processes. The pleiotropic benefits of this drug class derive from the extraordinary functionality of prolyl isomerization as a regulatory mechanism and its evolutionary diversification into many biochemical pathways. Nonimmunosuppressive analogs of cyclosporine A are the most thoroughly characterized cyclophilin inhibitors and show significant potential to attenuate several of the major pathophysiological events in NASH - mitochondrial dysfunction, cellular injury and death, inflammation, and in particular, fibrosis.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , Ciclofilinas/metabolismo , Ciclosporinas/farmacología , Ciclosporinas/uso terapéutico , Desarrollo de Medicamentos , Inhibidores Enzimáticos/farmacología , Humanos , Enfermedad del Hígado Graso no Alcohólico/fisiopatología
17.
Cells ; 8(11)2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31717385

RESUMEN

Hepatic fibrosis can result as a pathological response to nonalcoholic steatohepatitis (NASH). Cirrhosis, the late stage of fibrosis, has been linked to poor survival and an increased risk of developing hepatocellular carcinoma, with limited treatment options available. Therefore, there is an unmet need for novel effective antifibrotic compounds. Cyclophilins are peptidyl-prolyl cis-trans isomerases that facilitate protein folding and conformational changes affecting the function of the targeted proteins. Due to their activity, cyclophilins have been presented as key factors in several stages of the fibrotic process. In this study, we investigated the antifibrotic effects of NV556, a novel potent sanglifehrin-based cyclophilin inhibitor, in vitro and in vivo. NV556 potential antifibrotic effect was evaluated in two well-established animal models of NASH, STAM, and methionine-choline-deficient (MCD) mice, as well as in an in vitro 3D human liver ECM culture of LX2 cells, a human hepatic stellate cell line. We demonstrate that NV556 decreased liver fibrosis in both STAM and MCD in vivo models and decreased collagen production in TGFß1-activated hepatic stellate cells in vitro. Taken together, these results present NV556 as a potential candidate for the treatment of liver fibrosis.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Cirrosis Hepática/metabolismo , Animales , Deficiencia de Colina , Colágeno Tipo I/metabolismo , Dieta , Modelos Animales de Enfermedad , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Humanos , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Masculino , Metionina/deficiencia , Ratones , Terapia Molecular Dirigida , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología
18.
Bioorg Med Chem Lett ; 29(23): 126717, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31635932

RESUMEN

Fragment-based screening by SPR enabled the discovery of chemical diverse fragment hits with millimolar binding affinities to the peptidyl-prolyl isomerase Cyclophilin D (CypD). The CypD protein crystal structures of 6 fragment hits provided the basis for subsequent medicinal chemistry optimization by fragment merging and linking yielding three different chemical series with either urea, oxalyl or amide linkers connecting millimolar fragments in the S1' and S2 pockets. We successfully improved the in vitro CypD potencies in the biochemical FP and PPIase assays and in the biophysical SPR binding assay from millimolar towards the low micromolar and submicromolar range by >1000-fold for some fragment derivatives. The initial SAR together with the protein crystal structures of our novel CypD inhibitors provide a suitable basis for further hit-to-lead optimization.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Lactamas/farmacología , Cristalografía por Rayos X , Ciclofilinas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Lactamas/síntesis química , Lactamas/química , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
19.
J Pharmacol Exp Ther ; 371(2): 231-241, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31406003

RESUMEN

Previous studies show that cyclophilins contribute to many pathologic processes, and cyclophilin inhibitors demonstrate therapeutic activities in many experimental models. However, no drug with cyclophilin inhibition as the primary mode of action has advanced completely through clinical development to market. In this study, we present findings on the cyclophilin inhibitor, CRV431, that highlight its potential as a drug candidate for chronic liver diseases. CRV431 was found to potently inhibit all cyclophilin isoforms tested-A, B, D, and G. Inhibitory constant or IC50 values ranged from 1 to 7 nM, which was up to 13 times more potent than the parent compound, cyclosporine A (CsA), from which CRV431 was derived. Other CRV431 advantages over CsA as a nontransplant drug candidate were significantly diminished immunosuppressive activity, less drug transporter inhibition, and reduced cytotoxicity potential. Oral dosing to mice and rats led to good blood exposures and a 5- to 15-fold accumulation of CRV431 in liver compared with blood concentrations across a wide range of CRV431 dosing levels. Most importantly, CRV431 decreased liver fibrosis in a 6-week carbon tetrachloride model and in a mouse model of nonalcoholic steatohepatitis (NASH). Additionally, CRV431 administration during a late, oncogenic stage of the NASH disease model resulted in a 50% reduction in the number and size of liver tumors. These findings are consistent with CRV431 targeting fibrosis and cancer through multiple, cyclophilin-mediated mechanisms and support the development of CRV431 as a safe and effective drug candidate for liver diseases. SIGNIFICANCE STATEMENT: Cyclophilin inhibitors have demonstrated therapeutic activities in many disease models, but no drug candidates have yet advanced completely through development to market. In this study, CRV431 is shown to potently inhibit multiple cyclophilin isoforms, possess several optimized pharmacological properties, and decrease liver fibrosis and tumors in mouse models of chronic liver disease, which highlights its potential to be the first approved drug primarily targeting cyclophilin isomerases.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Ciclosporinas/uso terapéutico , Modelos Animales de Enfermedad , Enfermedad Hepática en Estado Terminal/tratamiento farmacológico , Cirrosis Hepática/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ciclosporinas/farmacología , Relación Dosis-Respuesta a Droga , Enfermedad Hepática en Estado Terminal/patología , Femenino , Humanos , Células Jurkat , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Ratas , Ratas Sprague-Dawley , Carga Tumoral/efectos de los fármacos , Carga Tumoral/fisiología
20.
PLoS One ; 14(6): e0217433, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31181107

RESUMEN

Hepatitis B virus (HBV) infection is a major health burden worldwide with 240 million chronically infected individuals. Nucleos(t)ide analogs and interferons are the current standards of care due to their suppression of HBV replication, but the treatments rarely eradicate HBV from individuals. Similar to current treatments for human immunodeficiency virus type-1 (HIV-1) and hepatitis C virus (HCV) patients, improved HBV therapies will require the combination of multiple drugs which target distinct steps of the HBV life cycle. In this study, we tested the potential of a cyclophilin inhibitor, CRV431, to affect HBV replication in transgenic mice. We found that oral treatment with CRV431 (50 mg/kg/day) for a period of 16 days significantly reduced liver HBV DNA levels and moderately decreased serum HBsAg levels. We observed an additive inhibitory effect on liver HBV DNA levels in mice treated with a combination of low doses of CRV431 (10 mg/kg/day) and the nucleotide prodrug, tenofovir exalidex (TXL), (5 mg/kg/day). No toxicity was observed in CRV431-treated mice. Although it is well known that CRV431 neutralizes the peptidyl-prolyl isomerase activity of cyclophilins, its anti-HBV mechanism(s) of action remains unknown. Nevertheless, this study provides the first demonstration of a beneficial effect of a cyclophilin inhibitor in vivo in an HBV transgenic mouse model. Altogether our data reveal the potential of CRV431 to be part of improved new therapies for HBV patients.


Asunto(s)
Antivirales/farmacología , Ciclofilinas/antagonistas & inhibidores , Ciclosporinas/farmacología , ADN Viral/efectos de los fármacos , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Animales , Femenino , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Hepatitis B/virología , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/virología , Hepatitis C/tratamiento farmacológico , Hepatitis C/virología , Humanos , Hígado/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nucleótidos/farmacología , Profármacos/farmacología , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA