Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 395
Filtrar
1.
Artículo en Alemán | MEDLINE | ID: mdl-37956666

RESUMEN

Feline infectious peritonitis (FIP) is one of the most common infectious diseases in cats that is fatal when untreated. So far, there is no legally available effective treatment in Germany. Treatment options include only symptomatic treatment (e. g. glucocorticoids, propentofylline), immunomodulatory approaches (e. g. interferons, polyprenyl immunostimulant), and antiviral chemotherapy with protease inhibitors (e. g. GC376) or nucleoside analogues (e. g. GS-441524, remdesivir). Symptomatic treatment does not cure FIP but may lead to a short-term improvement of clinical signs in a subset of cats. Immunomodulatory treatment has also not shown to be very promising. In contrary, the antiviral compounds GS-441524 and GC376 exhibited significant efficacy in several studies and their use saved the lives of many cats suffering from FIP. However, both agents are currently not licensed and thus cannot be legally administered by veterinarians in Germany. Legally, cats may only be legally treated with GS-441524 in a few countries (e.g. Great Britain and Australia). In other countries, GS-441524 is imported by cat owners via the black market and administered on their own. This article provides an overview of the available treatment options and an outlook on the legal use of effective antiviral drugs.


Asunto(s)
Enfermedades de los Gatos , Peritonitis Infecciosa Felina , Animales , Gatos , Antivirales/uso terapéutico , Enfermedades de los Gatos/tratamiento farmacológico , Peritonitis Infecciosa Felina/tratamiento farmacológico , Ácidos Sulfónicos/uso terapéutico , Resultado del Tratamiento
2.
Cell Host Microbe ; 30(8): 1074-1076, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35952645

RESUMEN

Tackling the ancient infectious foe of malaria, Xie et al. (2022) uncover a novel class of nucleoside analogs that selectively hijack and inhibit the tyrosine tRNA synthase of the parasite. With high potency, good oral bioavailability, and minimal host cell toxicity, these inhibitors show promise as next-generation antimalarials.


Asunto(s)
Antimaláricos , Malaria , Adenosina , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Humanos , Malaria/tratamiento farmacológico , Malaria/parasitología , Plasmodium falciparum/genética , Ácidos Sulfónicos/uso terapéutico
3.
J Tradit Chin Med ; 42(4): 565-575, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35848973

RESUMEN

OBJECTIVE: To investigate the therapeutic effect of Sishen Wan (, SSW) on ulcerative colitis (UC) induced by dinitrobenzene sulfonic acid and its effect on toll-like receptor 2/interleukin-1 receptor-associated kinase-4/nuclear factor-κB (TLR2/IRAK4/NF-κB) sig-naling pathway in colonic tissue. METHODS: In this study, 120 Sprague-Dawley rats were randomly divided into blank and model groups. The experimental UC model in rats was established by subcutaneous injection of hydrocortisone + senna gavage for 21 d + dinitrobenzene sulfonic acid (DNBS)/ ethanol solution enema. The successful model rats were randomly divided into the model group; mesalazine (0.36 g/kg) group; and high-, medium-, and low- dose SSW (24, 12, and 6 g/kg) groups. The model and blank groups were gavaged with equal volumes of distilled water once a day for 21 d. The general condition of the rats was observed, and the body mass, fecal properties, and occult blood were recorded for calculating the disease activity index (DAI) score. The colonic tissue of the rats was collected, and its general morphology and pathological form were noted for obtaining the colonic mucosal injury index (CMDI) score. Hematoxylin-eosin staining was used to view the pathological changes of the colon tissue in each group, apoptosis of the cells was detected using terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining, and quantitative real-time polymerase chain reaction was used to measure the expressions of TLR2, myeloid differentiation primary response gene 88 (MyD88), IRAK4, and NF-κB p65 mRNA in the colon tissue. The expressions of TLR2, MyD88, IRAK4, and NF-κB p65 protein were detected using western blotting and immunohistochemistry assay, and the levels of interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) in the colon tissue were determined using enzyme linked immunosorbent assay. RESULTS: Compared with the blank group, the general condition of the model group was relatively poor. The DAI and CMDI scores of the model group increased significantly (< 0.01), the glands and intestinal mucosa disappeared partially, and several inflammatory cells infiltrated and gathered in the mucosal layer and base layer of the rats in the model group. Furthermore, the cell apoptosis and expression levels of TLR2, MyD88, IRAK4, and NF-κB p65 mRNA and protein in the colon tissue of rats in the model group increased significantly (< 0.01). The levels of IL-1ß and TNF-α increased significantly in the colon tissue of rats in the model group (< 0.01). After treatment with SSW, compared with the model group, the general condition of the UC rats improved. Moreover, the DAI and CMDI scores of the UC rats decreased significantly (< 0.05), and the pathological changes in the colon tissue of the UC rats tended to be normal. The cell apoptosis and expression levels of TLR2, MyD88, IRAK4, and NF-κB p65 mRNA and protein in the colon tissue of the UC rats decreased gradually ( < 0.01), and the levels of IL-1ß and TNF-α decreased significantly (< 0.01). CONCLUSION: SSW can improve the general condition and alleviate the intestinal mucosal injury of UC model rats. Additionally, SSW can inhibit the TLR2/IRAK4/ NF-κB signaling pathway, but further studies are required to confirm it.


Asunto(s)
Colitis Ulcerosa , FN-kappa B , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/genética , Dinitrobencenos , Medicamentos Herbarios Chinos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/farmacología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Ácidos Sulfónicos/metabolismo , Ácidos Sulfónicos/farmacología , Ácidos Sulfónicos/uso terapéutico , Receptor Toll-Like 2/genética , Factor de Necrosis Tumoral alfa/metabolismo
4.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35163378

RESUMEN

Hypertension significantly increases the risk of cardiovascular disease. Currently, effective standard pharmacological treatment is available in the form of diuretics, ACE inhibitors, angiotensin II receptor blockers and calcium channel blockers. These all help to decrease blood pressure in hypertensive patients, each with their own mechanism. Recently, firibastat, a new first-in-class antihypertensive drug has been developed. Firibastat is a prodrug that when crossing the blood-brain barrier, is cleaved into two active EC33 molecules. EC33 is the active molecule that inhibits the enzyme aminopeptidase A. Aminopeptidase A converts angiotensin II to angiotensin III. Angiotensin III usually has three central mechanisms that increase blood pressure, so by inhibiting this enzyme activity, a decrease in blood pressure is seen. Firibastat is an antihypertensive drug that affects the brain renin angiotensin system by inhibiting aminopeptidase A. Clinical trials with firibastat have been performed in animals and humans. No severe adverse effects related to firibastat treatment have been reported. Results from studies show that firibastat is generally well tolerated and safe to use in hypertensive patients. The aim of this review is to investigate the current knowledge about firibastat in the treatment of hypertension.


Asunto(s)
Arterias/patología , Disulfuros/uso terapéutico , Hipertensión/tratamiento farmacológico , Ácidos Sulfónicos/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Disulfuros/química , Disulfuros/farmacocinética , Disulfuros/farmacología , Humanos , Profármacos/química , Profármacos/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacocinética , Ácidos Sulfónicos/farmacología
5.
Cell Mol Gastroenterol Hepatol ; 13(4): 1201-1221, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34920145

RESUMEN

BACKGROUND & AIMS: TOB1 is an anti-proliferative protein of Tob/BTG family and typically involved in the tumorigenesis and T cell activation. Although TOB1 is associated with T helper 17 cell-related autoimmunity, its role in modulating T cell-mediated immune responses in IBD remains poorly understood. Here, we explored its expression and the underlying mechanisms involved in the pathogenesis of inflammatory bowel disease (IBD). METHODS: TOB1 and ID2 expression in IBD patients was examined by quantitative real time polymerase chain reaction and immunohistochemistry. IBD CD4+ T cells were transfected with lentivirus expressing TOB1, ID2, TOB1 short hairpin RNA and ID2 short hairpin RNA, respectively, and Tob1-/-CD4+ T cells were transfected with lentivirus expressing Id2. Experimental colitis was established in Tob1-/- mice by trinitrobenzene sulfonic acid enema and in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells to further explore the role of Tob1 in intestinal mucosal inflammation. Splenic CD4+ T cells of Tob1-/- mice were sorted to determine transcriptome differences by RNA sequencing. RESULTS: TOB1 expression was decreased in inflamed mucosa and peripheral blood CD4+ T cells of IBD patients compared with healthy subjects. Overexpression of TOB1 downregulated IBD CD4+ T cells to differentiate into Th1/Th17 cells compared with control subjects. Severe colitis was observed in Tob1-/- mice through trinitrobenzene sulfonic acid enema or in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells, compared with control animals. RNA sequencing analysis revealed ID2 as functional target of TOB1 to inhibit IBD CD4+ T cell differentiation into Th1/Th17 cells. Mechanistically, TOB1 was associated with Smad4/5 to induce ID2 expression and restrain Th1/Th17 cell differentiation. CONCLUSIONS: TOB1 restrains intestinal mucosal inflammation through suppressing Th1/Th17 cell-mediated immune responses via the Smad4/5-ID2 pathway. It may serve as a novel therapeutic target for treatment of human IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Proteínas de Homeodominio/metabolismo , Humanos , Inflamación/patología , Enfermedades Inflamatorias del Intestino/patología , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Mucosa Intestinal/metabolismo , Activación de Linfocitos , Ratones , ARN Interferente Pequeño/metabolismo , Ácidos Sulfónicos/metabolismo , Ácidos Sulfónicos/uso terapéutico , Células TH1 , Células Th17/metabolismo , Células Th17/patología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
6.
Eur J Med Chem ; 224: 113721, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34365127

RESUMEN

Many pentacyclic triterpenoids show anti-cancer and anti-inflammatory properties. Recently, we detected a pronounced cytotoxicity and radiosensitivity of two betulinyl sulfamates in human breast cancer cells. Besides betulinic acid scaffold (BSBA-S), we synthesized several new sulfamate-coupled scaffolds from oleanolic acid (OSBA-S), ursolic acid (USBA-S), platanic acid (PSBA-S) and maslinic acid (MSBA-S). Highest cytotoxicity was monitored in breast cancer cell lines after MSBA-S treatment showing in SRB assays IC50 values between 3.7 µM and 5.8 µM. Other sulfamate/triterpene conjugates, however, were less cytotoxic holding IC50 values between 6.6 µM and >50 µM, respectively. MSBA-S-treated breast cancer cells displayed significantly reduced clonogenic survival and an increased rate of apoptosis as compared to the other conjugates. In addition, MSBA-S in combination with irradiation resulted in effects on radiosensitivity in MDA-MB-231 cells (DMF10 = 1.14). In particular, ROS formation was strongly assessed in MSBA-S-treated breast cancer cells. Our findings suggest that the sulfamate derivative of maslinic acid MSBA-S might be a new option for the radiation therapy in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Herbicidas/uso terapéutico , Ácidos Sulfónicos/uso terapéutico , Femenino , Herbicidas/farmacología , Humanos , Estructura Molecular , Ácidos Sulfónicos/farmacología
7.
J Med Chem ; 64(5): 2501-2520, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33631934

RESUMEN

SUMOylation is a reversible post-translational modification that regulates protein function through covalent attachment of small ubiquitin-like modifier (SUMO) proteins. The process of SUMOylating proteins involves an enzymatic cascade, the first step of which entails the activation of a SUMO protein through an ATP-dependent process catalyzed by SUMO-activating enzyme (SAE). Here, we describe the identification of TAK-981, a mechanism-based inhibitor of SAE which forms a SUMO-TAK-981 adduct as the inhibitory species within the enzyme catalytic site. Optimization of selectivity against related enzymes as well as enhancement of mean residence time of the adduct were critical to the identification of compounds with potent cellular pathway inhibition and ultimately a prolonged pharmacodynamic effect and efficacy in preclinical tumor models, culminating in the identification of the clinical molecule TAK-981.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Ácidos Sulfónicos/uso terapéutico , Sumoilación/efectos de los fármacos , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Humanos , Ratones , Estructura Molecular , Unión Proteica , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Relación Estructura-Actividad , Ácidos Sulfónicos/síntesis química , Ácidos Sulfónicos/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Eur J Nutr ; 60(5): 2537-2551, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33216193

RESUMEN

PURPOSE: Probiotics have been shown to exert beneficial effects in IBD although their exact mechanisms are not completely understood. The aim of the present study was to assess the intestinal anti-inflammatory activity of different probiotics (Lactobacillus fermentum CECT5716, Lactobacillus salivarius CECT5713, Escherichia coli Nissle 1917, Saccharomyces boulardii CNCMI-745 in the dinitrobenzene sulfonic acid (DNBS) model of mouse colitis and correlate it with the modifications of the gut microbiota and the immune response, focusing on miRNA expression. METHODS: The probiotics were daily administered orally for 25 days. On day 19 colitis was induced by rectal installation of DNBS. At the end of the treatment, mice were sacrificed and the colonic damage was assessed biochemically by analysing the expression of different markers involved in the immune response, including miRNAs; and the colonic microbiota by pyrosequencing. Probiotics properties were also evaluated in vitro in different immune cell types (CMT-93 intestinal epithelial cells and bone marrow-derived macrophages), where the expression of different mRNAs and miRNAs was examined. RESULTS: All the probiotics displayed intestinal anti-inflammatory effects but slightly different, especially regarding miRNAs expression. Likewise, the probiotics ameliorated the colitis-associated dysbiosis, although showing differences in the main bacterial groups affected. CONCLUSION: Among the probiotics assayed, Lactobacillus fermentum CECT5716 and Escherichia coli Nissle 1917 appear to present the best intestinal anti-inflammatory effects, being the latter one of the few probiotics with reputed efficacy in human IBD. Therefore, Lactobacillus fermentum CECT5716 could be considered as a complementary nutritional strategy for IBD treatment.


Asunto(s)
Colitis , Microbioma Gastrointestinal , MicroARNs , Probióticos , Animales , Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Colitis/terapia , Dinitrobencenos/uso terapéutico , Ratones , MicroARNs/genética , Ácidos Sulfónicos/uso terapéutico
9.
Cardiovasc Res ; 117(6): 1546-1556, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-32653904

RESUMEN

AIMS: Receptor-type vascular endothelial protein tyrosine phosphatase (VE-PTP) dephosphorylates Tie-2 as well as CD31, VE-cadherin, and vascular endothelial growth factor receptor 2 (VEGFR2). The latter form a signal transduction complex that mediates the endothelial cell response to shear stress, including the activation of the endothelial nitric oxide (NO) synthase (eNOS). As VE-PTP expression is increased in diabetes, we investigated the consequences of VE-PTP inhibition (using AKB-9778) on blood pressure in diabetic patients and the role of VE-PTP in the regulation of eNOS activity and vascular reactivity. METHODS AND RESULTS: In diabetic patients AKB-9778 significantly lowered systolic and diastolic blood pressure. This could be linked to elevated NO production, as AKB increased NO generation by cultured endothelial cells and elicited the NOS inhibitor-sensitive relaxation of endothelium-intact rings of mouse aorta. At the molecular level, VE-PTP inhibition increased the phosphorylation of eNOS on Tyr81 and Ser1177 (human sequence). The PIEZO1 activator Yoda1, which was used to mimic the response to shear stress, also increased eNOS Tyr81 phosphorylation, an effect that was enhanced by VE-PTP inhibition. Two kinases, i.e. abelson-tyrosine protein kinase (ABL)1 and Src were identified as eNOS Tyr81 kinases as their inhibition and down-regulation significantly reduced the basal and Yoda1-induced tyrosine phosphorylation and activity of eNOS. VE-PTP, on the other hand, formed a complex with eNOS in endothelial cells and directly dephosphorylated eNOS Tyr81 in vitro. Finally, phosphorylation of eNOS on Tyr80 (murine sequence) was found to be reduced in diabetic mice and diabetes-induced endothelial dysfunction (isolated aortic rings) was blunted by VE-PTP inhibition. CONCLUSIONS: VE-PTP inhibition enhances eNOS activity to improve endothelial function and decrease blood pressure indirectly, through the activation of Tie-2 and the CD31/VE-cadherin/VEGFR2 complex, and directly by dephosphorylating eNOS Tyr81. VE-PTP inhibition, therefore, represents an attractive novel therapeutic option for diabetes-induced endothelial dysfunction and hypertension.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Antihipertensivos/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Hipertensión/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/antagonistas & inhibidores , Ácidos Sulfónicos/uso terapéutico , Animales , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Humanos , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Transducción de Señal , Resultado del Tratamiento , Estados Unidos
10.
PLoS One ; 15(11): e0241448, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33151949

RESUMEN

Equine sarcoid (ES) is the most prevalent skin tumor in equids worldwide. Additionally, aging grey horses frequently suffer from equine malignant melanoma (EMM). Current local therapies targeting these skin tumors remain challenging. Therefore, more feasible topical treatment options should be considered. In order to develop a topical therapy against ES and EMM, betulinyl-bis-sulfamate and NVX-207, derivatives of the naturally occurring betulin and betulinic acid, respectively, were evaluated for their antiproliferative (crystal violet staining assay), cytotoxic (MTS assay) and apoptotic (AnnexinV staining, cell cycle investigations) effects on primary ES cells, EMM cells and equine dermal fibroblasts in vitro. The more potent derivative was assessed for its in vitro penetration and permeation on isolated equine skin within 30 min and 24 h using Franz-type diffusion cells and HPLC analysis. Betulinyl-bis-sulfamate and NVX-207 inhibited the proliferation and metabolism in ES cells, EMM cells and fibroblasts significantly (p < 0.001) in a time- and dose-dependent manner. NVX-207 had superior anticancer effects compared to betulinyl-bis-sulfamate. Both compounds led to the externalization of phosphatidylserines on the cell membrane and DNA fragmentation, demonstrating that the effective mode of action was apoptosis. After 48 h of treatment with NVX-207, the number of necrotic cells was less than 2% in all cell types. Detected amounts of NVX-207 in the different skin layers exceeded the half-maximal inhibitory concentrations calculated by far. Even though data obtained in vitro are auspicious, the results are not unconditionally applicable to the clinical situation. Consequently, in vivo studies are required to address the antitumoral effects of topically applied NVX-207 in ES and EMM patients.


Asunto(s)
Enfermedades de los Caballos/tratamiento farmacológico , Propanolaminas/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/veterinaria , Ácidos Sulfónicos/uso terapéutico , Triterpenos/administración & dosificación , Triterpenos/uso terapéutico , Administración Tópica , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dermis/patología , Difusión , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Caballos , Concentración 50 Inhibidora , Propanolaminas/farmacología , Ácidos Sulfónicos/farmacología , Triterpenos/farmacología
11.
J Mater Chem B ; 8(46): 10609-10615, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33136104

RESUMEN

In this work, an anionic conjugated polyelectrolyte (PCP-SO3K), in which the backbone contains alternating 4,4-bis-alkyl-4H-cyclopenta-[2,1-b;3,4-b']-dithiophene and benzene structural units and the charges are provided by pendant sulfonate groups, was synthesized. The ionic nature of PCP-SO3K renders it soluble in water, and PCP-SO3K aqueous solution exhibits good photostability, with two main absorbance bands centered at 490 nm and 837 nm before and after laser irradiation. Its NIR absorption in water, negligible photoluminescence and insignificant intersystem crossing endow PCP-SO3K with efficient photothermal therapy performance, and an effective photothermal conversion efficiency of 56.7% was realized. Thus, PCP-SO3K aqueous solution can be used as an effective photothermal agent for in vivo applications as its photoactivity can be triggered by NIR light and can convert laser energy into thermal energy in a water environment. Of particular importance is the fact that complete tumor remission without recurrence in 4T1 tumor-bearing mice was realized after intravenous injection of PCP-SO3K aqueous solution and laser irradiation (2.0 W cm-2, 808 nm). The results indicate that the application of anionic conjugated polyelectrolytes as photothermal agents in photothermal therapy provides a new platform for the design of photothermal agents for clinical cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Polielectrolitos/uso terapéutico , Ácidos Sulfónicos/uso terapéutico , Tiofenos/uso terapéutico , Animales , Antineoplásicos/efectos de la radiación , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Rayos Infrarrojos , Ratones Endogámicos BALB C , Terapia Fototérmica/métodos , Polielectrolitos/efectos de la radiación , Polielectrolitos/toxicidad , Ácidos Sulfónicos/efectos de la radiación , Ácidos Sulfónicos/toxicidad , Tiofenos/efectos de la radiación , Tiofenos/toxicidad
12.
Sci Rep ; 10(1): 20787, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33247195

RESUMEN

Post-traumatic osteoarthritis (PTOA) is a major cause which hinders patients from the recovery after intra-articular injuries or surgeries. Currently, no effective treatment is available. In this study, we showed that inhibition of the acute stage chondrocyte death is a promising strategy to mitigate the development of PTOA. Namely, we examined efficacies of Kyoto University Substance (KUS) 121, a valosin-containing protein modulator, for PTOA as well as its therapeutic mechanisms. In vivo, in a rat PTOA model by cyclic compressive loading, intra-articular treatments of KUS121 significantly improved the modified Mankin scores and reduced damaged-cartilage volumes, as compared to vehicle treatment. Moreover, KUS121 markedly reduced the numbers of TUNEL-, CHOP-, MMP-13-, and ADAMTS-5-positive chondrocytes in the damaged knees. In vitro, KUS121 rescued human articular chondrocytes from tunicamycin-induced cell death, in both monolayer culture and cartilage explants. It also significantly downregulated the protein or gene expression of ER stress markers, proinflammatory cytokines, and extracellular-matrix-degrading enzymes induced by tunicamycin or IL-1ß. Collectively, these results demonstrated that KUS121 protected chondrocytes from cell death through the inhibition of excessive ER stress. Therefore, KUS121 would be a new, promising therapeutic agent with a protective effect on the progression of PTOA.


Asunto(s)
Naftalenos/uso terapéutico , Osteoartritis/tratamiento farmacológico , Piridinas/uso terapéutico , Ácidos Sulfónicos/uso terapéutico , Proteína que Contiene Valosina/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Anciano , Animales , Apoptosis/efectos de los fármacos , Cartílago Articular/efectos de los fármacos , Cartílago Articular/lesiones , Cartílago Articular/metabolismo , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Humanos , Masculino , Osteoartritis/etiología , Osteoartritis/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Tunicamicina/toxicidad , Heridas y Lesiones/complicaciones
13.
Oncol Rep ; 44(6): 2364-2372, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33125119

RESUMEN

Aspartate/asparagine ß­hydroxylase (AspH) is a type II transmembrane protein that catalyzes the post­translational hydroxylation of definite aspartyl and asparaginyl residues in epidermal growth factor­like domains of substrates. In the last few decades, accumulating evidence has indicated that AspH expression is upregulated in numerous types of human malignant cancer and is associated with poor survival and prognosis. The AspH protein aggregates on the surface of tumor cells, which contributes to inducing tumor cell migration, infiltration and metastasis. However, small­molecule inhibitors targeting hydroxylase activity can markedly block these processes, both in vitro and in vivo. Immunization of tumor­bearing mice with a phage vaccine fused with the AspH protein can substantially delay tumor growth and progression. Additionally, AspH antigen­specific CD4+ and CD8+ T cells were identified in the spleen of tumor­bearing mice. Therefore, these agents may be used as novel strategies for cancer treatment. The present review summarizes the current progress on the underlying mechanisms of AspH expression in cancer development.


Asunto(s)
Antígenos de Neoplasias/genética , Proteínas de Unión al Calcio/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Proteínas de la Membrana/genética , Oxigenasas de Función Mixta/genética , Proteínas Musculares/genética , Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/inmunología , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Movimiento Celular/inmunología , Furanos/farmacología , Furanos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Oxigenasas de Función Mixta/antagonistas & inhibidores , Oxigenasas de Función Mixta/inmunología , Oxigenasas de Función Mixta/metabolismo , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/inmunología , Proteínas Musculares/metabolismo , Invasividad Neoplásica/genética , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/patología , Neoplasias/inmunología , Neoplasias/mortalidad , Neoplasias/patología , Pronóstico , Ácidos Sulfónicos/farmacología , Ácidos Sulfónicos/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Neuroreport ; 31(15): 1096-1103, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-32881774

RESUMEN

Our previous study showed that propane-2-sulfonic acid octadec-9-enyl-amide (N15), a novel peroxisome proliferator-activated receptor α and γ (PPARα/γ) dual agonist, inhibits inflammatory responses in tumor necrosis factor alpha (TNFα)-induced vascular endothelial cells or lipopolysaccharide (LPS)-induced human myeloid leukemia mononuclear cells-1. However, little is known about whether N15 applies to other pathological or neuroinflammatory conditions. In the present study, we detected the effect of N15 on the LPS-induced neuroinflammatory response in mice and further investigated whether the effect of N15 on neuroinflammation and neuronal cells survival was related to PPARα/γ dual pathways. We found that N15 decreased the mRNA expression of the proinflammatory cytokines IL-1ß, IL-6, TNFα, inducible nitric oxide synthase and cyclooxygenase-2; inhibited microglial activation; and ameliorated neuronal apoptosis in the hippocampus and cortex of LPS-induced mice. In addition, PPARα antagonist MK886 or PPARγ antagonist T0070907 partially eliminated the effect of N15. These results demonstrate that N15 exerts an anti-inflammatory effect, at least in part, by enhancing PPARα/γ dual signaling. Our study reveals that N15 may be a promising neuronal protective drug for the treatment of neuroinflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , PPAR alfa/agonistas , PPAR gamma/agonistas , Ácidos Sulfónicos/farmacología , Animales , Antiinflamatorios/uso terapéutico , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ácidos Sulfónicos/uso terapéutico
15.
Biochem Biophys Res Commun ; 529(2): 283-288, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32703424

RESUMEN

Our previous reports demonstrated that the novel peroxisome proliferator-activated receptors α and γ (PPARα/γ) dual agonist propane-2-sulfonic acid octadec-9-enyl-amide (N15) alleviates cognitive ability in the chronic phase of ischemic stroke. However, the potential effects of N15 on Alzheimer's disease (AD) animal models have not been elucidated. In the present study, we investigated the effects of N15 on scopolamine-induced cognitive dysfunction and cholinergic system ability. N15 (50, 100 or 200 mg/kg) was administered to mice via oral gavage for 21 days, and spatial cognitive dysfunction was induced via an intraperitoneal injection of scopolamine (4 mg/kg) for 6 days. We found that N15 pretreatment markedly ameliorated scopolamine-induced spatial cognitive impairment and enhanced cholinergic system reactivity in the hippocampus. N15 pretreatment also significantly increased the expression levels of growth-associated protein-43, synaptophysin, brain-derived neurotrophic factor and neurotrophin-3 in the hippocampus. Our data demonstrate that N15 has an anti-amnesic effect, which may be mediated by enhancing cholinergic activity and synaptic plasticity. These findings support N15 as a potent neuropharmacological drug against AD.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , PPAR alfa/agonistas , PPAR gamma/agonistas , Aprendizaje Espacial/efectos de los fármacos , Ácidos Sulfónicos/uso terapéutico , Animales , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Ratones , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Escopolamina
17.
Can J Cardiol ; 36(5): 721-731, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32389345

RESUMEN

The pathophysiology of heart failure (HF) and hypertension are thought to involve brain renin-angiotensin system (RAS) hyperactivity. Angiotensin III, a key effector peptide in the brain RAS, provides tonic stimulatory control over blood pressure (BP) in hypertensive rats. Aminopeptidase A (APA), the enzyme responsible for generating brain angiotensin III, constitutes a potential therapeutic target for hypertension treatment. We focus here on studies of RB150/firibastat, the first prodrug of the specific and selective APA inhibitor EC33 able to cross the blood-brain barrier. We consider its development from therapeutic target discovery to clinical trials of the prodrug. After oral administration, firibastat crosses the gastrointestinal and blood-brain barriers. On arrival in the brain, it is cleaved to generate EC33, which inhibits brain APA activity, lowering BP in various experimental models of hypertension. Firibastat was clinically and biologically well tolerated, even at high doses, in phase I trials conducted in healthy human subjects. It was then shown to decrease BP effectively in patients of various ethnic origins with hypertension in phase II trials. Brain RAS hyperactivity leads to excessive sympathetic activity, which can contribute to HF after myocardial infarction (MI). Chronic treatment with oral firibastat (4 or 8 weeks after MI) has been shown to normalize brain APA activity in mice. This effect is accompanied by a normalization of brain RAS and sympathetic activities, reducing cardiac fibrosis and hypertrophy and preventing cardiac dysfunction. Firibastat may therefore represent a novel therapeutic advance in the clinical management of patients with hypertension and potentially with HF after MI.


Asunto(s)
Encéfalo/metabolismo , Glutamil Aminopeptidasa/antagonistas & inhibidores , Insuficiencia Cardíaca/prevención & control , Hipertensión/tratamiento farmacológico , Angiotensina II/metabolismo , Angiotensina III/metabolismo , Animales , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico , Ensayos Clínicos como Asunto , Disulfuros/farmacología , Disulfuros/uso terapéutico , Glutamil Aminopeptidasa/metabolismo , Insuficiencia Cardíaca/etiología , Humanos , Hipertensión/metabolismo , Infarto del Miocardio/complicaciones , Sistema Renina-Angiotensina , Ácidos Sulfónicos/farmacología , Ácidos Sulfónicos/uso terapéutico
18.
Cancer Lett ; 469: 78-88, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31629931

RESUMEN

Selective estrogen receptor modulators (SERMs) are a class of structurally diverse compounds, which have been extensively used to treat hormone-responsive cancers due to their unique partially agonistic and antagonistic properties toward estrogen receptors. Our previous studies have identified a three-dimensional SERM, oxabicycloheptene sulfonate (OBHS), as an estrogen receptor α (ERα) ligand, which is effective for the prevention and treatment of estrogen-dependent endometriosis in vivo. Here, using genome-wide ChIP-seq and RNA-seq analysis, we report that OBHS rapidly induces genome-wide ERα occupancy and acts as a partial agonist and antagonist for ERα. Interestingly, OBHS downregulates the homologous recombination and repair (HRR) modules, resulting in increased DNA damage, apoptosis and cell cycle arrest, inducing synthetic lethality with poly (ADP-ribose) polymerase (PARP) inhibitor olaparib through ERα antagonism. Mechanistically, OBHS impairs the RNA polymerase II (Pol II) loading at the promoters of estrogen-responsive HRR genes. Furthermore, combination therapy of OBHS with olaparib significantly reduces the tumour burden and delays the progression of breast cancer in vivo. Together, our studies not only characterise a novel SERM which uniquely targets the homologous recombination and repair programmes through ERα antagonism but also propose a synthetic lethal strategy by combining OBHS with PARP inhibitor olaparib for ERα-responsive cancers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Antagonistas del Receptor de Estrógeno/farmacología , Reparación del ADN por Recombinación/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Ácidos Sulfónicos/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Chlorocebus aethiops , Secuenciación de Inmunoprecipitación de Cromatina , Antagonistas del Receptor de Estrógeno/uso terapéutico , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Ratones , Estructura Molecular , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/antagonistas & inhibidores , ARN Polimerasa II/metabolismo , RNA-Seq , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Relación Estructura-Actividad , Ácidos Sulfónicos/uso terapéutico , Mutaciones Letales Sintéticas/efectos de los fármacos , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Mol Sci ; 20(23)2019 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-31810330

RESUMEN

Carbonic anhydrases (CAs) are a family of enzymes involved in the pH regulation of metabolically active cells/tissues. Upregulation of the CAIX/XII isoforms is associated with hypoxic tumours and clinically linked with malignant progression, treatment resistance and poor prognosis. The elucidation of the crystal structure of the catalytic domains of CAIX/XII provided the basis for the generation of CAIX/XII selective inhibitors based on the sulfonamide, sulfamate and coumarins chemical structures. Ureido-substituted benzenesulfonamide CAIX/XII inhibitors have shown significant potential, with U-104 (SLC-0111) currently present in clinical Phase I/II. Ureido-substituted sulfamate CAIX/XII inhibitors have received less attention despite encouraging preclinical test results. In triple-negative breast cancer (TNBC), ureidosulfamates revealed a significant antitumour (FC9-398A) and antimetastatic potential (S4). In small cell lung cancer (SCLC), a cancer cell type very sensitive to a dysregulation in CAIX signaling, S4 treatment was particularly effective when combined with cisplatin with no evidence of acquired cisplatin-resistance. These successful anticancer strategies should provide a solid basis for future studies on ureido-substituted sulfamates.


Asunto(s)
Inhibidores de Anhidrasa Carbónica/uso terapéutico , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Relación Estructura-Actividad , Sulfonamidas/uso terapéutico , Anhidrasa Carbónica IX/antagonistas & inhibidores , Anhidrasa Carbónica IX/genética , Anhidrasas Carbónicas/efectos de los fármacos , Anhidrasas Carbónicas/genética , Humanos , Compuestos de Fenilurea/uso terapéutico , Ácidos Sulfónicos/química , Ácidos Sulfónicos/uso terapéutico
20.
Sci Rep ; 9(1): 11519, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31395936

RESUMEN

Reduced adenosine triphosphate (ATP) levels in ischemic stroke constitute an upstream contributor to neuronal cell death. We have recently created a small chemical, named Kyoto University Substance 121 (KUS121), which can reduce cellular ATP consumption. In this study, we examined whether KUS121 has neuroprotective effects in rodent cerebral ischemia models. We evaluated cell viability and ATP levels in vitro after oxygen glucose deprivation (OGD) in rat cortical primary neuronal cultures incubated with or without KUS121. We found that KUS121 protected neurons from cell death under OGD by preventing ATP depletion. We also used in vivo ischemic stroke models of transient distal middle cerebral artery occlusion in C57BL/6 and B-17 mice. Administration of KUS121 in these models improved functional deficits and reduced brain infarction volume after transient focal cerebral ischemia in both C57BL/6 and B-17 mice. These results indicate that KUS121 could be a novel type of neuroprotective drug for ischemic stroke.


Asunto(s)
Adenosina Trifosfato/metabolismo , Isquemia Encefálica/prevención & control , Naftalenos/farmacología , Fármacos Neuroprotectores/farmacología , Piridinas/farmacología , Accidente Cerebrovascular/prevención & control , Ácidos Sulfónicos/farmacología , Animales , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Infarto de la Arteria Cerebral Media , Ratones , Ratones Endogámicos C57BL , Naftalenos/uso terapéutico , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Piridinas/uso terapéutico , Reproducibilidad de los Resultados , Ácidos Sulfónicos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...