Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 568
Filtrar
1.
Biochem Pharmacol ; 224: 116252, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701866

RESUMEN

The mitogen-activated protein kinase (MAPK/ERK) pathway is pivotal in controlling the proliferation and survival of melanoma cells. Several mutations, including those in BRAF, exhibit an oncogenic effect leading to increased cellular proliferation. As a result, the combination therapy of a MEK inhibitor with a BRAF inhibitor demonstrated higher efficacy and lower toxicity than BRAF inhibitor alone. This combination has become the preferred standard of care for tumors driven by BRAF mutations. Aldehyde dehydrogenase 1A1 (ALDH1A1) is a known marker of stemness involved in drug resistance in several type of tumors, including melanoma. This study demonstrates that melanoma cells overexpressing ALDH1A1 displayed resistance to vemurafenib and trametinib through the activation of PI3K/AKT signaling instead of MAPK axis. Inhibition of PI3K/AKT signaling partially rescued sensitivity to the drugs. Consistently, pharmacological inhibition of ALDH1A1 activity downregulated the activation of AKT and partially recovered responsiveness to vemurafenib and trametinib. We propose ALDH1A1 as a new potential target for treating melanoma resistant to MAPK/ERK inhibitors.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1 , Resistencia a Antineoplásicos , Melanoma , Células Madre Neoplásicas , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas c-akt , Retinal-Deshidrogenasa , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Melanoma/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Línea Celular Tumoral , Familia de Aldehído Deshidrogenasa 1/metabolismo , Familia de Aldehído Deshidrogenasa 1/genética , Retinal-Deshidrogenasa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Pirimidinonas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Piridonas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vemurafenib/farmacología , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/genética , Antineoplásicos/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fenotipo
2.
Life Sci ; 320: 121541, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-36870386

RESUMEN

Modern cancer chemotherapy originated in the 1940s, and since then, many chemotherapeutic agents have been developed. However, most of these agents show limited response in patients due to innate and acquired resistance to therapy, which leads to the development of multi-drug resistance to different treatment modalities, leading to cancer recurrence and, eventually, patient death. One of the crucial players in inducing chemotherapy resistance is the aldehyde dehydrogenase (ALDH) enzyme. ALDH is overexpressed in chemotherapy-resistant cancer cells, which detoxifies the generated toxic aldehydes from chemotherapy, preventing the formation of reactive oxygen species and, thus, inhibiting the induction of oxidative stress and the stimulation of DNA damage and cell death. This review discusses the mechanisms of chemotherapy resistance in cancer cells promoted by ALDH. In addition, we provide detailed insight into the role of ALDH in cancer stemness, metastasis, metabolism, and cell death. Several studies investigated targeting ALDH in combination with other treatments as a potential therapeutic regimen to overcome resistance. We also highlight novel approaches in ALDH inhibition, including the potential synergistic employment of ALDH inhibitors in combination with chemotherapy or immunotherapy against different cancers, including head and neck, colorectal, breast, lung, and liver.


Asunto(s)
Aldehído Deshidrogenasa , Resistencia a Antineoplásicos , Inmunoterapia , Neoplasias , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/radioterapia , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Animales , Metástasis de la Neoplasia , Muerte Celular , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de la radiación
3.
Chem Commun (Camb) ; 58(1): 72-75, 2021 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-34874378

RESUMEN

Aldehyde dehydrogenase (ALDH), a cancer stem cell biomarker, is related to drug resistance. Co-treatment of anti-cancer drug (CPT) and ALDH inhibitor (DEAB) can overcome the drug resistance of cancer stem cells (CSCs) and finally cure cancers without relapse. We herein introduce a prodrug (DE-CPT) - consisting of 1,3-oxathiolane as an ROS responsive scaffold, and an aldehyde protecting group of DEAB - to deliver the CPT and DEAB upon reaction with ROS. From tests of the sphere-forming ability and CSC marker subpopulation, we found that DE-CPT efficiently decreases the CSCs population and kills the cancer cells.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Profármacos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Tiofenos/farmacología , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Células Madre Neoplásicas/metabolismo , Profármacos/química , Profármacos/metabolismo , Tiofenos/química , Tiofenos/metabolismo
4.
Molecules ; 26(21)2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34770988

RESUMEN

Glyceryl trinitrate (GTN) is one of the earliest known treatments for angina with a fascinating history that bridges three centuries. However, despite its central role in the nitric oxide (NO) story as a NO-donating compound, establishing the precise mechanism of how GTN exerts its medicinal benefit has proven to be far more difficult. This review brings together the explosive and vasodilatory nature of this three-carbon molecule while providing an update on the likely in vivo pathways through which GTN, and the rest of the organic nitrate family, release NO, nitrite, or a combination of both, while also trying to explain nitrate tolerance. Over the last 20 years the alcohol detoxification enzyme, aldehyde dehydrogenase (ALDH), has undoubtedly emerged as the front runner to explaining GTN's bioactivation. This is best illustrated by reduced GTN efficacy in subjects carrying the single point mutation (Glu504Lys) in ALDH, which is also responsible for alcohol intolerance, as characterized by flushing. While these findings are significant for anyone following the GTN story, they appear particularly relevant for healthcare professionals, and especially so, if administering GTN to patients as an emergency treatment. In short, although the GTN puzzle has not been fully solved, clinical study data continue to cement the importance of ALDH, as uncovered in 2002, as a key GTN activator.


Asunto(s)
Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Alcoholismo/tratamiento farmacológico , Aldehído Deshidrogenasa/antagonistas & inhibidores , Nitroglicerina/farmacología , Vasodilatadores/farmacología , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/metabolismo , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Animales , Humanos
5.
Toxicology ; 462: 152947, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34534558

RESUMEN

Phthalate plasticizers are commonly used in various consumer-end products. Human salivary aldehyde dehydrogenase (hsALDH) is a detoxifying enzyme which defends us from the toxic aldehydes. Here, the effect of phthalates [Di-2-ethylhexyl phthalate (DEHP), Diethyl phthalate (DEP) and Dibutyl phthalate (DBP)] on hsALDH has been investigated. These plasticizers inhibited hsALDH, and the IC50 values were 0.48 ± 0.04, 283.20 ± 0.09 and 285.00 ± 0.14 µM for DEHP, DEP and DBP, respectively. DEHP was the most potent inhibitor among the three plasticizers. They exhibited mixed-type linear inhibition with inclination towards competitive-non-competitive inhibition. They induced both tertiary and secondary structural changes in the enzyme. Quenching of intrinsic hsALDH fluorescence in a constant manner was observed with a binding constant (Kb) of 8.91 × 106, 2.80 × 104, and 1.31 × 105 M-1, for DEHP, DEP and DBP, respectively. Computational analysis showed that these plasticizers bind stably in the proximity of hsALDH catalytic site, reciprocating via non-covalent interactions with some of the amino acids which are evolutionary conserved. Therefore, exposure to these plasticizers inhibits hsALDH which increases the risk of aldehyde induced toxicity, adversely affecting oral health. The study has implications in assessing the safety of packaged food items which utilize phthalates.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Dibutil Ftalato/toxicidad , Ácidos Ftálicos/toxicidad , Plastificantes/toxicidad , Adulto , Dibutil Ftalato/administración & dosificación , Dietilhexil Ftalato/administración & dosificación , Dietilhexil Ftalato/toxicidad , Humanos , Concentración 50 Inhibidora , Ácidos Ftálicos/administración & dosificación , Plastificantes/administración & dosificación , Saliva/efectos de los fármacos , Saliva/enzimología
6.
Toxicol Appl Pharmacol ; 426: 115642, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34242567

RESUMEN

Disulfiram (DSF), a sulfur-containing compound, has been used to treat chronic alcoholism and cancer for decades by inactivating aldehyde dehydrogenase (ALDH). Hydrogen sulfide (H2S) is a new gasotransmitter and regulates various cellular functions by S-sulfhydrating cysteine in the target proteins. H2S exhibits similar properties to DSF in the sensitization of cancer cells. The interaction of DSF and H2S on ALDH activity and liver cancer cell survival are not clear. Here it was demonstrated that DSF facilitated H2S release from thiol-containing compounds, and DSF and H2S were both capable of regulating ALDH through inhibition of gene expression and enzymatic activity. The supplement of H2S sensitized human liver cancer cells (HepG2) to DSF-inhibited cell viability. The expression of cystathionine gamma-lyase (a major H2S-generating enzyme) was lower but ALDH was higher in mouse liver cancer stem cells (Dt81Hepa1-6) in comparison with their parental cells (Hepa1-6), and H2S was able to inhibit liver cancer stem cell adhesion. In conclusion, these data point to the potential of combining DSF and H2S for inhibition of cancer cell growth and tumor development by targeting ALDH.


Asunto(s)
Inhibidores del Acetaldehído Deshidrogenasa/farmacología , Disuasivos de Alcohol/farmacología , Aldehído Deshidrogenasa/antagonistas & inhibidores , Antineoplásicos/farmacología , Disulfiram/farmacología , Sulfuro de Hidrógeno/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Aldehído Deshidrogenasa/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cobre/farmacología , Humanos , Concentración de Iones de Hidrógeno , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Temperatura
7.
Ther Deliv ; 12(8): 611-625, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34286601

RESUMEN

Aim: The overexpression of aldehyde dehydrogenase (ALDH) in cancer cells contributes to therapeutic resistance. Furazolidone (FUR) is a strong ALDH inhibitor. Methods: FUR nanoemulsion (NE) was formulated and tested for ALDH inhibitory activity in comparison with free FUR. The cytotoxic potential of cisplatin was evaluated in combination with free FUR and FUR NE. Results: The optimized FUR NE showed droplet size of 167.9 ± 3.1 nm and drug content of 84.2 ± 2.3%. FUR NE inhibited 99.75 ± 2.1% of ALDH activity while 25.0 ± 4.6% was inhibited by free FUR. FUR NE increased the sensitivity to cisplatin in A549 cells by more than tenfold by its ALDH inhibitory effects. Conclusion: This finding can be a promising approach to improve cancer survival in ALDH-positive drug-resistant cancers.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Cisplatino , Furazolidona/farmacología , Neoplasias Pulmonares , Células A549 , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Nanoestructuras
8.
J Pharmacol Exp Ther ; 379(1): 33-40, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34321315

RESUMEN

As the central regulator of the oxidative stress response, nuclear factor erythroid 2-related factor 2 (Nrf2) is attracting great interest as a therapeutic target for various cancers, and the possible clinical applications of novel Nrf2 inhibitors have been explored in Nrf2-activated cancers. In the present study, we specifically investigated halofuginone, which is derived from a natural plant alkaloid. We found that halofuginone administration decreased the number of pancreatic intraepithelial neoplasias in pancreas-specific Kras and p53 mutant (KPC) mice. In Nrf2-activated pancreatic cancer cell lines established from KPC mice, halofuginone rapidly depleted Nrf2 in Nrf2-activated cancer cells. Both in vitro and in vivo, it sensitized Nrf2-activated pancreatic cancer cells to gemcitabine, which is the first-line chemotherapy in clinical practice. In our mechanistic study, we found that halofuginone downregulated aldehyde dehydrogenase 3a1 (ALDH3A1) in mouse pancreatic cancer cells. The Nrf2 inducer diethyl maleate upregulated ALDH3A1, and knockdown of Aldh3a1 sensitized Nrf2-activated cancer cells to gemcitabine, strongly suggesting that ALDH3A1 is regulated by Nrf2 and that it contributes to gemcitabine resistance. The current study demonstrated the therapeutic benefits of halofuginone in Nrf2-activated pancreatic cancers. SIGNIFICANCE STATEMENT: We identified nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target aldehyde dehydrogenase 3a1 (ALDH3A1) as novel therapeutic targets in pancreatic cancer. They negatively affect the efficacy of a conventional chemotherapeutic agent, gemcitabine. We confirmed that Nrf2 plays a pivotal role in the induction of ALDH3A1.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Desoxicitidina/análogos & derivados , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/metabolismo , Aldehído Deshidrogenasa/antagonistas & inhibidores , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Piperidinas/farmacología , Piperidinas/uso terapéutico , Quinazolinonas/farmacología , Quinazolinonas/uso terapéutico , Gemcitabina
9.
Am J Ophthalmol ; 226: 22-31, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33529588

RESUMEN

PURPOSE: To assess the activity of reproxalap, a novel reactive aldehyde species (RASP) inhibitor, relative to vehicle in patients with dry eye disease (DED) DESIGN: Randomized, double-masked, vehicle-controlled Phase 2b trial METHODS: Three hundred patients with DED were randomly assigned 1:1:1 at multiple US sites to receive 0.1% topical ocular reproxalap, 0.25% topical ocular reproxalap, or vehicle. Eyes were treated bilaterally 4 times daily for 12 weeks. Standard signs and symptoms of DED were assessed at baseline and at Weeks 2, 4, 8, and 12. RESULTS: A dose response was observed for signs and symptoms of DED. Relative to vehicle over 12 weeks of therapy, the largest symptomatic improvement was observed in ocular dryness (0.25%, P = .047), and the largest objective sign improvement was observed in nasal region fluorescein staining (0.25%, P = .030). A greater proportion of patients receiving 0.25% reproxalap vs. vehicle reported dryness scores of 0 (P = .012). Improvements in combined DED symptoms were evident by the first post-baseline visit (Week 2, 0.25%, P < .0001) in patients with baseline scores greater than or equal to median values. No significant changes in safety measures were observed. CONCLUSION: The novel RASP inhibitor reproxalap demonstrated rapid, broad, and clinically relevant symptomatic control, in conjunction with statistically significant improvement over vehicle in signs of DED as demonstrated by fluorescein staining, in DED patients over 12 weeks of therapy. The results represent the first vehicle-controlled evidence for the therapeutic potential of RASP inhibition to mitigate the signs and symptoms of dry eye disease.


Asunto(s)
Aminoquinolinas/uso terapéutico , Síndromes de Ojo Seco/tratamiento farmacológico , Administración Oftálmica , Anciano , Aldehído Deshidrogenasa/antagonistas & inhibidores , Córnea/metabolismo , Córnea/fisiopatología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Síndromes de Ojo Seco/diagnóstico , Síndromes de Ojo Seco/fisiopatología , Femenino , Fluoresceína/metabolismo , Colorantes Fluorescentes/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Soluciones Oftálmicas , Coloración y Etiquetado/métodos , Lágrimas/fisiología , Resultado del Tratamiento
10.
Molecules ; 26(2)2021 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-33429981

RESUMEN

Resident cancer cells with stem cell-like features induce drug tolerance, facilitating survival of glioblastoma (GBM). We previously showed that strategies targeting tumor bioenergetics present a novel emerging avenue for treatment of GBM. The objective of this study was to enhance the therapeutic effects of dual inhibition of tumor bioenergetics by combination of gossypol, an aldehyde dehydrogenase inhibitor, and phenformin, a biguanide compound that depletes oxidative phosphorylation, with the chemotherapeutic drug, temozolomide (TMZ), to block proliferation, stemness, and invasiveness of GBM tumorspheres (TSs). Combination therapy with gossypol, phenformin, and TMZ induced a significant reduction in ATP levels, cell viability, stemness, and invasiveness compared to TMZ monotherapy and dual therapy with gossypol and phenformin. Analysis of differentially expressed genes revealed up-regulation of genes involved in programmed cell death, autophagy, and protein metabolism and down-regulation of those associated with cell metabolism, cycle, and adhesion. Combination of TMZ with dual inhibitors of tumor bioenergetics may, therefore, present an effective strategy against GBM by enhancing therapeutic effects through multiple mechanisms of action.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Glioblastoma , Proteínas de Neoplasias/antagonistas & inhibidores , Esferoides Celulares/enzimología , Aldehído Deshidrogenasa/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/enzimología , Complejo I de Transporte de Electrón/metabolismo , Inhibidores Enzimáticos/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/enzimología , Humanos , Proteínas de Neoplasias/metabolismo , Temozolomida/farmacología
11.
Int J Med Sci ; 18(1): 53-64, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390773

RESUMEN

Mulberry leaves (Morus alba L.), which are traditional Chinese herbs, exert several biological functions, such as antioxidant, anti-inflammation, antidiabetic, and antitumor. Alcohol intake increases inflammation and oxidative stress, and this increase causes liver injury and leads to liver steatosis, cirrhosis, and hepatocellular carcinoma, which are major health problems worldwide. Previous report indicated that mulberry leaf extract (MLE) exited hepatoprotection effects against chronic alcohol-induced liver damages. In this present study, we investigated the effects of MLE on acute alcohol and liver injury induced by its metabolized compound called acetaldehyde (ACE) by using in vivo and in vitro models. Administration of MLE reversed acute alcohol-induced liver damages, increased acetaldehyde (ACE) level, and decreased aldehyde dehydrogenase activity in a dose-dependent manner. Acute alcohol exposure-induced leukocyte infiltration and pro-inflammation factors, including cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6), were blocked by MLE in proportion to MLE concentration. MLE prevented alcohol-induced liver apoptosis via enhanced caveolin-1 expression and attenuated EGFR/STAT3/iNOS pathway using immunohistochemical analysis. ACE induced proteins, such as iNOS, COX-2, TNF-α, and IL-6, and inhibited superoxide dismutase expression, whereas co-treated with MLE reversed these proteins expression. MLE also recovered alcohol-induced apoptosis in cultured Hep G2 cells. Overall, our findings indicated that MLE ameliorated acute alcohol-induced liver damages by reducing ACE toxicity and inhibiting apoptosis caused by oxidative stress signals. Our results implied that MLE might be a potential agent for treating alcohol liver disease.


Asunto(s)
Acetaldehído/toxicidad , Antioxidantes/administración & dosificación , Hepatopatías Alcohólicas/tratamiento farmacológico , Morus/química , Extractos Vegetales/administración & dosificación , Acetaldehído/metabolismo , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/metabolismo , Animales , Antioxidantes/aislamiento & purificación , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Pruebas de Enzimas , Etanol/administración & dosificación , Etanol/efectos adversos , Etanol/metabolismo , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Hepatopatías Alcohólicas/etiología , Hepatopatías Alcohólicas/patología , Ratones , Ratones Endogámicos ICR , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/aislamiento & purificación , Hojas de la Planta/química , Especies Reactivas de Oxígeno/metabolismo
12.
Eur J Med Chem ; 211: 113060, 2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33341649

RESUMEN

There is strong evidence that inhibition of one or more Aldehyde Dehydrogenase 1A (ALDH1A) isoforms may be beneficial in chemotherapy-resistant ovarian cancer and other tumor types. While many previous efforts have focused on development of ALDH1A1 selective inhibitors, the most deadly ovarian cancer subtype, high-grade serous (HGSOC), exhibits elevated expression of ALDH1A3. Herein, we report continued development of pan-ALDH1A inhibitors to assess whether broad spectrum ALDH1A inhibition is an effective adjunct to chemotherapy in this critical tumor subtype. Optimization of the CM39 scaffold, aided by metabolite ID and several new ALDH1A1 crystal structures, led to improved biochemical potencies, improved cellular ALDH inhibition in HGSOC cell lines, and substantial improvements in microsomal stability culminating in orally bioavailable compounds. We demonstrate that two compounds 68 and 69 are able to synergize with chemotherapy in a resistant cell line and patient-derived HGSOC tumor spheroids, indicating their suitability for future in vivo proof of concept experiments.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Aldehído Deshidrogenasa/farmacología , Femenino , Humanos , Estructura Molecular , Relación Estructura-Actividad
13.
PLoS Genet ; 16(9): e1009040, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32970669

RESUMEN

Genetic hearing loss is a common health problem with no effective therapy currently available. DFNA15, caused by mutations of the transcription factor POU4F3, is one of the most common forms of autosomal dominant non-syndromic deafness. In this study, we established a novel mouse model of the human DFNA15 deafness, with a Pou4f3 gene mutation (Pou4f3Δ) identical to that found in a familial case of DFNA15. The Pou4f3(Δ/+) mice suffered progressive deafness in a similar manner to the DFNA15 patients. Hair cells in the Pou4f3(Δ/+) cochlea displayed significant stereociliary and mitochondrial pathologies, with apparent loss of outer hair cells. Progression of hearing and outer hair cell loss of the Pou4f3(Δ/+) mice was significantly modified by other genetic and environmental factors. Using Pou4f3(-/+) heterozygous knockout mice, we also showed that DFNA15 is likely caused by haploinsufficiency of the Pou4f3 gene. Importantly, inhibition of retinoic acid signaling by the aldehyde dehydrogenase (Aldh) and retinoic acid receptor inhibitors promoted Pou4f3 expression in the cochlear tissue and suppressed the progression of hearing loss in the mutant mice. These data demonstrate Pou4f3 haploinsufficiency as the main underlying cause of human DFNA15 deafness and highlight the therapeutic potential of Aldh inhibitors for treatment of progressive hearing loss.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Células Ciliadas Auditivas/patología , Pérdida Auditiva/tratamiento farmacológico , Pérdida Auditiva/etiología , Proteínas de Homeodominio/genética , Factor de Transcripción Brn-3C/genética , Animales , Benzaldehídos/farmacología , Modelos Animales de Enfermedad , Haploinsuficiencia/genética , Pérdida Auditiva/genética , Pérdida Auditiva/patología , Proteínas de Homeodominio/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ruido/efectos adversos , Quinolinas/farmacología , Factor de Transcripción Brn-3C/metabolismo , Tretinoina/farmacología , para-Aminobenzoatos/farmacología
14.
Eur J Pharmacol ; 886: 173541, 2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-32896553

RESUMEN

Chronic alcoholism (CA) decreases bone mass and increases the risk of hip fracture. Alcohol and its main metabolite, acetaldehyde impairs osteoblastogenesis by increasing oxidative stress. Aldehyde dehydrogenase (ALDH) is the rate-limiting enzyme in clearing acetaldehyde from the body. The clinical relevance of ALDH in skeletal function has been established by the discovery of single nucleotide polymorphism, SNP (rs671) in the ALDH2 gene giving rise to an inactive form of the enzyme (ALDH2*2) that causes increased serum acetaldehyde and osteoporosis in the affected individuals. Subsequent mouse genetics studies have replicated human phenotype in mice and confirmed the non-redundant role of ALDH2 in bone homeostasis. The activity of ALDH2 is amenable to pharmacological modulation. ALDH2 inhibition by disulfiram (DSF) and activation by alda-1 cause reduction and induction of bone formation, respectively. DSF also inhibits peak bone mass accrual in growing rats. On the other hand, DSF showed an anti-osteoclastogenic effect and protected mice from alcohol-induced osteopenia by inhibiting ALDH1a1 in bone marrow monocytes. Besides DSF, there are several classes of ALDH inhibitors with disparate skeletal effects. Alda-1, the ALDH2 activator induced osteoblast differentiation by increasing bone morphogenic protein 2 (BMP2) expression via ALDH2 activation. Alda-1 also restored ovariectomy-induced bone loss. The scope of structure-activity based studies with ALDH2 and the alda-1-like molecule could lead to the discovery of novel osteoanabolic molecules. This review will critically discuss the molecular mechanism of the ethanol and its principal metabolite, acetaldehyde in the context of ALDH2 in bone cells, and skeletal homeostasis.


Asunto(s)
Aldehído Deshidrogenasa/efectos de los fármacos , Enfermedades Óseas/tratamiento farmacológico , Alcoholismo/complicaciones , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/genética , Aldehídos/metabolismo , Animales , Enfermedades Óseas/etiología , Etanol/metabolismo , Humanos , Osteogénesis/efectos de los fármacos
15.
Cells ; 9(9)2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32883024

RESUMEN

The greatest challenge in cancer therapy is posed by drug-resistant recurrence following treatment. Anticancer chemotherapy is largely focused on targeting the rapid proliferation and biosynthesis of cancer cells. This strategy has the potential to trigger autophagy, enabling cancer cell survival through the recycling of molecules and energy essential for biosynthesis, leading to drug resistance. Autophagy recycling contributes amino acids and ATP to restore mTOR complex 1 (mTORC1) activity, which leads to cell survival. However, autophagy with mTORC1 activation can be stalled by reducing the ATP level. We have previously shown that cytosolic NADH production supported by aldehyde dehydrogenase (ALDH) is critical for supplying ATP through oxidative phosphorylation (OxPhos) in cancer cell mitochondria. Inhibitors of the mitochondrial complex I of the OxPhos electron transfer chain and ALDH significantly reduce the ATP level selectively in cancer cells, terminating autophagy triggered by anticancer drug treatment. With the aim of overcoming drug resistance, we investigated combining the inhibition of mitochondrial complex I, using phenformin, and ALDH, using gossypol, with anticancer drug treatment. Here, we show that OxPhos targeting combined with anticancer drugs acts synergistically to enhance the anticancer effect in mouse xenograft models of various cancers, which suggests a potential therapeutic approach for drug-resistant cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Gosipol/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Fenformina/uso terapéutico , Aldehído Deshidrogenasa/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Sinergismo Farmacológico , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Gosipol/farmacología , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias/patología , Fenformina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Adv Exp Med Biol ; 1258: 157-166, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32767240

RESUMEN

Aldehyde dehydrogenases are a family of enzymes that oxidize aldehydes to carboxylic acids. These enzymes are important in cellular homeostasis during oxidative stress by the elimination of toxic aldehyde by-products from various cellular processes. In osteosarcoma, aldehyde dehydrogenase 1A1has been described as a cancer stem cell marker. Its activity has been found to correlate with metastatic potential and the metastatic phenotype. As such, a more complete understanding of aldehyde dehydrogenase in osteosarcoma will give us a deeper knowledge of its impact on osteosarcoma metastatic potential. Our hope is that this knowledge can be translated into novel antimetastatic therapeutic strategies and thus improve osteosarcoma prognoses.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Osteosarcoma/tratamiento farmacológico , Aldehídos/metabolismo , Neoplasias Óseas/enzimología , Humanos , Células Madre Neoplásicas/enzimología , Osteosarcoma/enzimología
17.
Arch Biochem Biophys ; 691: 108477, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32717224

RESUMEN

Aldehyde dehydrogenase 9A1 (ALDH9A1) is a human enzyme that catalyzes the NAD+-dependent oxidation of the carnitine precursor 4-trimethylaminobutyraldehyde to 4-N-trimethylaminobutyrate. Here we show that the broad-spectrum ALDH inhibitor diethylaminobenzaldehyde (DEAB) reversibly inhibits ALDH9A1 in a time-dependent manner. Possible mechanisms of inhibition include covalent reversible inactivation involving the thiohemiacetal intermediate and slow, tight-binding inhibition. Two crystal structures of ALDH9A1 are reported, including the first of the enzyme complexed with NAD+. One of the structures reveals the active conformation of the enzyme, in which the Rossmann dinucleotide-binding domain is fully ordered and the inter-domain linker adopts the canonical ß-hairpin observed in other ALDH structures. The oligomeric structure of ALDH9A1 was investigated using analytical ultracentrifugation, small-angle X-ray scattering, and negative stain electron microscopy. These data show that ALDH9A1 forms the classic ALDH superfamily dimer-of-dimers tetramer in solution. Our results suggest that the presence of an aldehyde substrate and NAD+ promotes isomerization of the enzyme into the active conformation.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/química , Aldehído Deshidrogenasa/metabolismo , Benzaldehídos/química , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Humanos , Cinética , NAD/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína
18.
J Clin Pharmacol ; 60(12): 1598-1605, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32578227

RESUMEN

ANS-6637, a pro-drug of GS-548351, is a selective, reversible inhibitor of aldehyde dehydrogenase isoform 2 under development as an anticraving agent for the treatment of substance use disorders. In vitro testing indicates that GS-548351 is an inhibitor and inducer of cytochrome P450 family 3, subfamily A (CYP3A). In this phase 1 single-center, open-label, fixed-sequence drug-drug interaction study we assessed the impact of steady-state GS-548351 on single-dose pharmacokinetics of midazolam, an index substrate for CYP3A. Twelve healthy volunteers received 600 mg of ANS-6637 by mouth daily from study days 3 to 8 and a single 5-mg oral dose of midazolam on days 1 and 8. Pharmacokinetic samples were collected over 24 hours on days 1 and 8, then analyzed using liquid chromatography-tandem mass spectrometry. The prespecified no-effect range for the 90% confidence interval (CI) of the geometric mean ratio (GMR) of midazolam coadministered with ANS-6637 (day 8) compared with midazolam alone (day 1) was 0.7-1.43. There was an increase in midazolam AUC0-∞ (GMR [90%CI]) that was within the no-effect range (1.26 [1.12-1.425]) and an increase in midazolam Cmax that was outside the range (1.22 [1.03-1.45]). The AUC0-∞ (1.08 [0.91-1.27]) and Cmax (0.95 [0.75-1.2]) of 1-hydroxymidazolam, the primary metabolite of midazolam, were also within the no-effect range. A single grade 3 adverse event (alanine aminotransferase elevation) was identified and resolved following discontinuation of the study drug. Overall, multidose ANS-6637 was well tolerated and did not alter the PK of midazolam beyond a small increase in AUC0-∞ that is unlikely to be clinically significant.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Midazolam/farmacocinética , Compuestos Orgánicos/farmacología , Profármacos/farmacología , Administración Oral , Adulto , Aldehído Deshidrogenasa/antagonistas & inhibidores , Área Bajo la Curva , Citocromo P-450 CYP3A/metabolismo , Esquema de Medicación , Interacciones Farmacológicas , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/sangre , Femenino , Semivida , Voluntarios Sanos , Humanos , Masculino , Midazolam/administración & dosificación , Midazolam/análogos & derivados , Midazolam/sangre , Midazolam/metabolismo , Compuestos Orgánicos/administración & dosificación , Profármacos/administración & dosificación , Profármacos/efectos adversos , Profármacos/metabolismo
19.
Oncogene ; 39(13): 2756-2771, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32015486

RESUMEN

Aldehyde dehydrogenases (ALDHs) are multifunctional enzymes that oxidize diverse endogenous and exogenous aldehydes. We conducted a meta-analysis based on The Cancer Genome Atlas and Gene Expression Omnibus data and detected genetic alterations in ALDH1A1, ALDH1A3, or ALDH3A1, 86% of which were gene amplification or mRNA upregulation, in 31% of nonsmall cell lung cancers (NSCLCs). The expression of these isoenzymes impacted chemoresistance and shortened survival times in patients. We hypothesized that these enzymes provide an oxidative advantage for the persistence of NSCLC. To test this hypothesis, we used genetic and pharmacological approaches with DIMATE, an irreversible inhibitor of ALDH1/3. DIMATE showed cytotoxicity in 73% of NSCLC cell lines tested and demonstrated antitumor activity in orthotopic xenografts via hydroxynonenal-protein adduct accumulation, GSTO1-mediated depletion of glutathione and increased H2O2. Consistent with this result, ALDH1/3 disruption synergized with ROS-inducing agents or glutathione synthesis inhibitors to trigger cell death. In lung cancer xenografts with high to moderate cisplatin resistance, combination treatment with DIMATE promoted strong synergistic responses with tumor regression. These results indicate that NSCLCs with increased expression of ALDH1A1, ALDH1A3, or ALDH3A1 may be targeted by strategies involving inhibitors of these isoenzymes as monotherapy or in combination with chemotherapy to overcome patient-specific drug resistance.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/antagonistas & inhibidores , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Oxidorreductasas/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Retinal-Deshidrogenasa/antagonistas & inhibidores , Anciano , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1/genética , Familia de Aldehído Deshidrogenasa 1/metabolismo , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Alquinos/farmacología , Alquinos/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Amplificación de Genes , Glutatión/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Ratones , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Compuestos de Sulfhidrilo/farmacología , Compuestos de Sulfhidrilo/uso terapéutico , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Chembiochem ; 21(13): 1911-1917, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31985142

RESUMEN

Aldehyde dehydrogenases (ALDHs) convert aldehydes into carboxylic acids and are often upregulated in cancer. They have been linked to therapy resistance and are therefore potential therapeutic targets. However, only a few selective and potent inhibitors are currently available for this group of enzymes. Competitive activity-based protein profiling (ABPP) would aid the development and validation of new selective inhibitors. Herein, a broad-spectrum activity-based probe that reports on several ALDHs is presented. This probe was used in a competitive ABPP protocol against three ALDH inhibitors in lung cancer cells to determine their selectivity profiles and establish their target engagement.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Inhibidores Enzimáticos/química , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/genética , Línea Celular Tumoral , Química Clic , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Colorantes Fluorescentes/química , Humanos , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...