Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 572
Filter
1.
Life Sci ; 356: 123033, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39222837

ABSTRACT

Human ALDH comprise 19 subfamilies in which ALDH1A1, ALDH1A3, ALDH3A1, ALDH5A1, ALDH7A1, and ALDH18A1 are implicated in CSC. Studies have shown that ALDH can also be involved in drug resistance and standard chemotherapy regimens are ineffective in treating patients at the stage of disease recurrence. Existing chemotherapeutic drugs eliminate the bulk of tumors but are usually not effective against CSC which express ALDH+ population. Henceforth, targeting ALDH is convincing to treat the patient's post-relapse. Combination therapies that interlink signaling mechanisms seem promising to increase the overall disease-free survival rate. Therefore, targeting ALDH through ALDH inhibitors along with immunotherapies may create a novel platform for translational research. This review aims to fill in the gap between ALDH1 family members in relation to its cell signaling mechanisms, highlighting their potential as molecular targets to sensitize recurrent tumors and bring forward the future development concerning the current progress and draw backs. This review summarizes the role of cancer stem cells and their upregulation by maintaining the tumor microenvironment in which ALDH is specifically highlighted. It discusses the regulation of ALDH family proteins and the crosstalk between ALDH and CSC in relation to cancer metabolism. Furthermore, it establishes the correlation between ALDH involved signaling mechanisms and their specific targeted inhibitors, as well as their functional modularity, bioavailability, and mechanistic role in various cancers.


Subject(s)
Aldehyde Dehydrogenase , Neoplasms , Neoplastic Stem Cells , Tumor Microenvironment , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Tumor Microenvironment/drug effects , Molecular Targeted Therapy , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
J Agric Food Chem ; 72(35): 19424-19435, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39172074

ABSTRACT

Fusarium verticillioides is the primary pathogen causing ear rot and stalk rot in corn (Zea mays). It not only affects yields but also produces mycotoxins endangering both human and animal health. Aldehyde dehydrogenase (ALDH) is essential for the oxidation of aldehydes in living organisms, making it a potential target for human drug design. However, there are limited reports on its function in plant pathogenic fungus. In this study, we analyzed the expression levels and gene knockout mutants, revealing that ALDH genes FvALDH-43 and FvALDH-96 in F. verticillioides played significant roles in pathogenicity and resistance to low-temperature stress by affecting antioxidant capacity. Virtual screening for natural product inhibitors and molecular docking were performed targeting FvALDH-43 and FvALDH-96. Following the biological activity analysis, three natural flavonoid compounds featuring a 2-hydroxyphenol chromene were identified. Among these, Taxifolin exhibited the highest biological activity and low toxicity. Both in vitro and in vivo biological evaluations confirmed that Taxifolin targeted ALDH and inhibited its activity. These findings indicate that aldehyde dehydrogenase may serve as a promising target for the design of novel fungicides.


Subject(s)
Aldehyde Dehydrogenase , Fungal Proteins , Fungicides, Industrial , Fusarium , Molecular Docking Simulation , Plant Diseases , Zea mays , Fusarium/enzymology , Fusarium/genetics , Fusarium/drug effects , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/chemistry , Fungicides, Industrial/pharmacology , Fungicides, Industrial/chemistry , Fungal Proteins/genetics , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungal Proteins/antagonists & inhibitors , Zea mays/microbiology , Zea mays/chemistry , Plant Diseases/microbiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry
3.
Molecules ; 29(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38999066

ABSTRACT

Aldehyde dehydrogenases (ALDHs) are a family of enzymes that aid in detoxification and are overexpressed in several different malignancies. There is a correlation between increased expression of ALDH and a poor prognosis, stemness, and resistance to several drugs. Several ALDH inhibitors have been generated due to the crucial role that ALDH plays in cancer stem cells. All of these inhibitors, however, are either ineffective, very toxic, or have yet to be subjected to rigorous testing on their effectiveness. Although various drug-like compounds targeting ALDH have been reported in the literature, none have made it to routine use in the oncology clinic. As a result, new potent, non-toxic, bioavailable, and therapeutically effective ALDH inhibitors are still needed. In this study, we designed and synthesized potent multi-ALDH isoform inhibitors based on the isatin and indazole pharmacophore. Molecular docking studies and enzymatic tests revealed that among all of the synthesized analogs, compound 3 is the most potent inhibitor of ALDH1A1, ALDH3A1, and ALDH1A3, exhibiting 51.32%, 51.87%, and 36.65% inhibition, respectively. The ALDEFLUOR assay further revealed that compound 3 acts as an ALDH broad spectrum inhibitor at 500 nM. Compound 3 was also the most cytotoxic to cancer cells, with an IC50 in the range of 2.1 to 3.8 µM for ovarian, colon, and pancreatic cancer cells, compared to normal and embryonic kidney cells (IC50 7.1 to 8.7 µM). Mechanistically, compound 3 increased ROS activity due to potent multi-ALDH isoform inhibition, which increased apoptosis. Taken together, this study identified a potent multi-isoform ALDH inhibitor that could be further developed as a cancer therapeutic.


Subject(s)
Aldehyde Dehydrogenase , Enzyme Inhibitors , Isatin , Molecular Docking Simulation , Humans , Isatin/chemistry , Isatin/pharmacology , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Structure
4.
Biochem Pharmacol ; 224: 116252, 2024 06.
Article in English | MEDLINE | ID: mdl-38701866

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Drug Resistance, Neoplasm , Melanoma , Neoplastic Stem Cells , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt , Retinal Dehydrogenase , Humans , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Cell Line, Tumor , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Retinal Dehydrogenase/metabolism , Protein Kinase Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pyrimidinones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Pyridones/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Vemurafenib/pharmacology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/genetics , Antineoplastic Agents/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Phenotype
5.
Naunyn Schmiedebergs Arch Pharmacol ; 397(9): 6963-6973, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38613572

ABSTRACT

Levofloxacin (LVX) is among the fluoroquinolones antibiotics that has also been studied in vitro and in vivo for its anticancer effects. In this study, we used LVX and novel LVX thionated derivatives; compounds 2 and 3, to evaluate their antioxidant activity, aldehyde dehydrogenase (ALDH) enzymes activity inhibition, and anticancer activity. Combination treatments with doxorubicin (DOX) were investigated as well. The 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay was used to determine the antioxidant activity. The NADH fluorescence spectrophotometric activity assay was used to determine the ALDH inhibitory effects. Resazurin dye method was applied for cell viability assays. Molecular Operating Environment software was used for the molecular docking experiments. Compared to ascorbic acid, DPPH assay showed that compound 3 had the highest antioxidant activity among the tested compounds with approximately 35% scavenging activity. On ALDH enzymes, compound 3 showed a significant ALDH activity inhibition compared to compound 2 at 200 µM. The IC50 values for the tested compounds were approximately 100 µM on A549 cell line, a non-small cell lung cancer (NSCLC) cell line. However, significant enhancement of cytotoxicity and reduction of IC50 values were observed by combining DOX and synergism was achieved with LVX with a combination index value of 0.4. The molecular docking test showed a minimum binding energy with a good affinity for compound 3 towards ALDH enzymes. Thionated LVX derivatives, may be repurposed for NSCLC therapy in combination with DOX, taking into account the antioxidant activity, ALDH activity inhibition, and the molecular docking results of compound 3.


Subject(s)
Aldehyde Dehydrogenase , Antioxidants , Cell Survival , Doxorubicin , Levofloxacin , Molecular Docking Simulation , Levofloxacin/pharmacology , Humans , A549 Cells , Antioxidants/pharmacology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Cell Survival/drug effects , Doxorubicin/pharmacology , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Biphenyl Compounds/pharmacology
6.
Life Sci ; 320: 121541, 2023 May 01.
Article in English | MEDLINE | ID: mdl-36870386

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase , Drug Resistance, Neoplasm , Immunotherapy , Neoplasms , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Drug Resistance, Neoplasm/drug effects , Humans , Animals , Neoplasm Metastasis , Cell Death , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects
7.
Chem Commun (Camb) ; 58(1): 72-75, 2021 Dec 21.
Article in English | MEDLINE | ID: mdl-34874378

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism , Thiophenes/pharmacology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Neoplastic Stem Cells/metabolism , Prodrugs/chemistry , Prodrugs/metabolism , Thiophenes/chemistry , Thiophenes/metabolism
8.
Molecules ; 26(21)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34770988

ABSTRACT

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.


Subject(s)
Alcohol Drinking/drug therapy , Alcoholism/drug therapy , Aldehyde Dehydrogenase/antagonists & inhibitors , Nitroglycerin/pharmacology , Vasodilator Agents/pharmacology , Alcohol Drinking/metabolism , Alcoholism/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Animals , Humans
9.
Toxicology ; 462: 152947, 2021 10.
Article in English | MEDLINE | ID: mdl-34534558

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Dibutyl Phthalate/toxicity , Phthalic Acids/toxicity , Plasticizers/toxicity , Adult , Dibutyl Phthalate/administration & dosage , Diethylhexyl Phthalate/administration & dosage , Diethylhexyl Phthalate/toxicity , Humans , Inhibitory Concentration 50 , Phthalic Acids/administration & dosage , Plasticizers/administration & dosage , Saliva/drug effects , Saliva/enzymology
10.
Toxicol Appl Pharmacol ; 426: 115642, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34242567

ABSTRACT

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.


Subject(s)
Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Alcohol Deterrents/pharmacology , Aldehyde Dehydrogenase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Disulfiram/pharmacology , Hydrogen Sulfide/metabolism , Liver Neoplasms/drug therapy , Aldehyde Dehydrogenase/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Copper/pharmacology , Humans , Hydrogen-Ion Concentration , Liver/drug effects , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Temperature
11.
J Pharmacol Exp Ther ; 379(1): 33-40, 2021 10.
Article in English | MEDLINE | ID: mdl-34321315

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/therapeutic use , Cell Line, Tumor , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-E2-Related Factor 2/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Piperidines/pharmacology , Piperidines/therapeutic use , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Gemcitabine
12.
Ther Deliv ; 12(8): 611-625, 2021 08.
Article in English | MEDLINE | ID: mdl-34286601

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Cisplatin , Furazolidone/pharmacology , Lung Neoplasms , A549 Cells , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Humans , Lung Neoplasms/drug therapy , Nanostructures
13.
Am J Ophthalmol ; 226: 22-31, 2021 06.
Article in English | MEDLINE | ID: mdl-33529588

ABSTRACT

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.


Subject(s)
Aminoquinolines/therapeutic use , Dry Eye Syndromes/drug therapy , Administration, Ophthalmic , Aged , Aldehyde Dehydrogenase/antagonists & inhibitors , Cornea/metabolism , Cornea/physiopathology , Dose-Response Relationship, Drug , Double-Blind Method , Dry Eye Syndromes/diagnosis , Dry Eye Syndromes/physiopathology , Female , Fluorescein/metabolism , Fluorescent Dyes/metabolism , Humans , Male , Middle Aged , Ophthalmic Solutions , Staining and Labeling/methods , Tears/physiology , Treatment Outcome
14.
Int J Med Sci ; 18(1): 53-64, 2021.
Article in English | MEDLINE | ID: mdl-33390773

ABSTRACT

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.


Subject(s)
Acetaldehyde/toxicity , Antioxidants/administration & dosage , Liver Diseases, Alcoholic/drug therapy , Morus/chemistry , Plant Extracts/administration & dosage , Acetaldehyde/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Animals , Antioxidants/isolation & purification , Apoptosis/drug effects , Disease Models, Animal , Enzyme Assays , Ethanol/administration & dosage , Ethanol/adverse effects , Ethanol/metabolism , Hep G2 Cells , Humans , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/pathology , Mice , Mice, Inbred ICR , Oxidative Stress/drug effects , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Reactive Oxygen Species/metabolism
15.
Molecules ; 26(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33429981

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms , Electron Transport Complex I/antagonists & inhibitors , Glioblastoma , Neoplasm Proteins/antagonists & inhibitors , Spheroids, Cellular/enzymology , Aldehyde Dehydrogenase/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Electron Transport Complex I/metabolism , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Glioblastoma/enzymology , Humans , Neoplasm Proteins/metabolism , Temozolomide/pharmacology
16.
Eur J Med Chem ; 211: 113060, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33341649

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/therapeutic use , Ovarian Neoplasms/drug therapy , Aldehyde Dehydrogenase/pharmacology , Female , Humans , Molecular Structure , Structure-Activity Relationship
17.
Eur J Pharmacol ; 886: 173541, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-32896553

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/drug effects , Bone Diseases/drug therapy , Alcoholism/complications , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/genetics , Aldehydes/metabolism , Animals , Bone Diseases/etiology , Ethanol/metabolism , Humans , Osteogenesis/drug effects
18.
PLoS Genet ; 16(9): e1009040, 2020 09.
Article in English | MEDLINE | ID: mdl-32970669

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/pathology , Hearing Loss/drug therapy , Hearing Loss/etiology , Homeodomain Proteins/genetics , Transcription Factor Brn-3C/genetics , Animals , Benzaldehydes/pharmacology , Disease Models, Animal , Haploinsufficiency/genetics , Hearing Loss/genetics , Hearing Loss/pathology , Homeodomain Proteins/metabolism , Humans , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Noise/adverse effects , Quinolines/pharmacology , Transcription Factor Brn-3C/metabolism , Tretinoin/pharmacology , para-Aminobenzoates/pharmacology
19.
Cells ; 9(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32883024

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Gossypol/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidative Phosphorylation/drug effects , Phenformin/therapeutic use , Aldehyde Dehydrogenase/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Drug Synergism , Electron Transport Complex I/antagonists & inhibitors , Gossypol/pharmacology , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/pathology , Phenformin/pharmacology , Xenograft Model Antitumor Assays
20.
Adv Exp Med Biol ; 1258: 157-166, 2020.
Article in English | MEDLINE | ID: mdl-32767240

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , Aldehydes/metabolism , Bone Neoplasms/enzymology , Humans , Neoplastic Stem Cells/enzymology , Osteosarcoma/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL