Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 300(5): 107271, 2024 May.
Article in English | MEDLINE | ID: mdl-38588813

ABSTRACT

Lafora disease (LD) is an autosomal recessive myoclonus epilepsy with onset in the teenage years leading to death within a decade of onset. LD is characterized by the overaccumulation of hyperphosphorylated, poorly branched, insoluble, glycogen-like polymers called Lafora bodies. The disease is caused by mutations in either EPM2A, encoding laforin, a dual specificity phosphatase that dephosphorylates glycogen, or EMP2B, encoding malin, an E3-ubiquitin ligase. While glycogen is a widely accepted laforin substrate, substrates for malin have been difficult to identify partly due to the lack of malin antibodies able to detect malin in vivo. Here we describe a mouse model in which the malin gene is modified at the C-terminus to contain the c-myc tag sequence, making an expression of malin-myc readily detectable. Mass spectrometry analyses of immunoprecipitates using c-myc tag antibodies demonstrate that malin interacts with laforin and several glycogen-metabolizing enzymes. To investigate the role of laforin in these interactions we analyzed two additional mouse models: malin-myc/laforin knockout and malin-myc/LaforinCS, where laforin was either absent or the catalytic Cys was genomically mutated to Ser, respectively. The interaction of malin with partner proteins requires laforin but is not dependent on its catalytic activity or the presence of glycogen. Overall, the results demonstrate that laforin and malin form a complex in vivo, which stabilizes malin and enhances interaction with partner proteins to facilitate normal glycogen metabolism. They also provide insights into the development of LD and the rescue of the disease by the catalytically inactive phosphatase.


Subject(s)
Lafora Disease , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin-Protein Ligases , Lafora Disease/metabolism , Lafora Disease/genetics , Lafora Disease/pathology , Animals , Mice , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Humans , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/genetics , Disease Models, Animal , Glycogen/metabolism , Glycogen/genetics
2.
Chem Biol Interact ; 391: 110910, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38364885

ABSTRACT

Aldehyde dehydrogenase 1A (ALDH1A) isoforms may be a useful target for overcoming chemotherapy resistance in high-grade serous ovarian cancer (HGSOC) and other solid tumor cancers. However, as different cancers express different ALDH1A isoforms, isoform selective inhibitors may have a limited therapeutic scope. Furthermore, resistance to an ALDH1A isoform selective inhibitor could arise via induction of expression of other ALDH1A isoforms. As such, we have focused on the development of pan-ALDH1A inhibitors, rather than on ALDH1A isoform selective compounds. Herein, we report the development of a new group of pan-ALDH1A inhibitors to assess whether broad spectrum ALDH1A inhibition is an effective adjunct to chemotherapy in HGSOC. Optimization of the CM10 scaffold, aided by ALDH1A1 crystal structures, led to improved biochemical potencies, improved cellular efficacy as demonstrated by reduction in ALDEFLUOR signal in HGSOC cells, and substantial improvements in liver microsomal stability. Based on this work we identified two compounds 17 and 25 suitable for future in vivo proof of concept experiments.


Subject(s)
Isoenzymes , Neoplasms , Humans , Aldehyde Dehydrogenase/metabolism , Retinal Dehydrogenase/metabolism , Aldehyde Oxidoreductases/metabolism
3.
Sci Transl Med ; 16(730): eadf1691, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38232139

ABSTRACT

Glycogen synthase 1 (GYS1), the rate-limiting enzyme in muscle glycogen synthesis, plays a central role in energy homeostasis and has been proposed as a therapeutic target in multiple glycogen storage diseases. Despite decades of investigation, there are no known potent, selective small-molecule inhibitors of this enzyme. Here, we report the preclinical characterization of MZ-101, a small molecule that potently inhibits GYS1 in vitro and in vivo without inhibiting GYS2, a related isoform essential for synthesizing liver glycogen. Chronic treatment with MZ-101 depleted muscle glycogen and was well tolerated in mice. Pompe disease, a glycogen storage disease caused by mutations in acid α glucosidase (GAA), results in pathological accumulation of glycogen and consequent autophagolysosomal abnormalities, metabolic dysregulation, and muscle atrophy. Enzyme replacement therapy (ERT) with recombinant GAA is the only approved treatment for Pompe disease, but it requires frequent infusions, and efficacy is limited by suboptimal skeletal muscle distribution. In a mouse model of Pompe disease, chronic oral administration of MZ-101 alone reduced glycogen buildup in skeletal muscle with comparable efficacy to ERT. In addition, treatment with MZ-101 in combination with ERT had an additive effect and could normalize muscle glycogen concentrations. Biochemical, metabolomic, and transcriptomic analyses of muscle tissue demonstrated that lowering of glycogen concentrations with MZ-101, alone or in combination with ERT, corrected the cellular pathology in this mouse model. These data suggest that substrate reduction therapy with GYS1 inhibition may be a promising therapeutic approach for Pompe disease and other glycogen storage diseases.


Subject(s)
Glycogen Storage Disease Type II , Mice , Animals , Glycogen Storage Disease Type II/drug therapy , Glycogen Synthase/metabolism , Glycogen Synthase/pharmacology , Mice, Knockout , Glycogen/metabolism , Muscle, Skeletal/metabolism , Enzyme Replacement Therapy/methods
4.
Cancers (Basel) ; 14(14)2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35884498

ABSTRACT

Ovarian cancer is a deadly disease attributed to late-stage detection as well as recurrence and the development of chemoresistance. Ovarian cancer stem cells (OCSCs) are hypothesized to be largely responsible for the emergence of chemoresistant tumors. Although chemotherapy may initially succeed at decreasing the size and number of tumors, it leaves behind residual malignant OCSCs. In this study, we demonstrate that aldehyde dehydrogenase 1A1 (ALDH1A1) is essential for the survival of OCSCs. We identified a first-in-class ALDH1A1 inhibitor, compound 974, and used 974 as a tool to decipher the mechanism of stemness regulation by ALDH1A1. The treatment of OCSCs with 974 significantly inhibited ALDH activity, the expression of stemness genes, and spheroid and colony formation. An in vivo limiting dilution assay demonstrated that 974 significantly inhibited CSC frequency. A transcriptomic sequencing of cells treated with 974 revealed a significant downregulation of genes related to stemness and chemoresistance as well as senescence and the senescence-associated secretory phenotype (SASP). We confirmed that 974 inhibited the senescence and stemness induced by platinum-based chemotherapy in functional assays. Overall, these data establish that ALDH1A1 is essential for OCSC survival and that ALDH1A1 inhibition suppresses chemotherapy-induced senescence and stemness. Targeting ALDH1A1 using small-molecule inhibitors in combination with chemotherapy therefore presents a promising strategy to prevent ovarian cancer recurrence and has the potential for clinical translation.

5.
Cell Rep ; 40(1): 111041, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35793618

ABSTRACT

Glycogen is the primary energy reserve in mammals, and dysregulation of glycogen metabolism can result in glycogen storage diseases (GSDs). In muscle, glycogen synthesis is initiated by the enzymes glycogenin-1 (GYG1), which seeds the molecule by autoglucosylation, and glycogen synthase-1 (GYS1), which extends the glycogen chain. Although both enzymes are required for proper glycogen production, the nature of their interaction has been enigmatic. Here, we present the human GYS1:GYG1 complex in multiple conformations representing different functional states. We observe an asymmetric conformation of GYS1 that exposes an interface for close GYG1 association, and propose this state facilitates handoff of the GYG1-associated glycogen chain to a GYS1 subunit for elongation. Full activation of GYS1 widens the GYG1-binding groove, enabling GYG1 release concomitant with glycogen chain growth. This structural mechanism connecting chain nucleation and extension explains the apparent stepwise nature of glycogen synthesis and suggests distinct states to target for GSD-modifying therapeutics.


Subject(s)
Glycogen Synthase , Glycogenolysis , Glycoproteins , Glucosyltransferases/metabolism , Glycogen/metabolism , Glycogen Synthase/metabolism , Glycoproteins/metabolism , Humans
6.
Cell Metab ; 33(7): 1404-1417.e9, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34043942

ABSTRACT

Glycosylation defects are a hallmark of many nervous system diseases. However, the molecular and metabolic basis for this pathology is not fully understood. In this study, we found that N-linked protein glycosylation in the brain is metabolically channeled to glucosamine metabolism through glycogenolysis. We discovered that glucosamine is an abundant constituent of brain glycogen, which functions as a glucosamine reservoir for multiple glycoconjugates. We demonstrated the enzymatic incorporation of glucosamine into glycogen by glycogen synthase, and the release by glycogen phosphorylase by biochemical and structural methodologies, in primary astrocytes, and in vivo by isotopic tracing and mass spectrometry. Using two mouse models of glycogen storage diseases, we showed that disruption of brain glycogen metabolism causes global decreases in free pools of UDP-N-acetylglucosamine and N-linked protein glycosylation. These findings revealed fundamental biological roles of brain glycogen in protein glycosylation with direct relevance to multiple human diseases of the central nervous system.


Subject(s)
Brain/metabolism , Glucosamine/metabolism , Glycogen/physiology , Protein Processing, Post-Translational , Animals , Cells, Cultured , Disease Models, Animal , Female , Glycogen/metabolism , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Glycogenolysis/genetics , Glycosylation , Lafora Disease/genetics , Lafora Disease/metabolism , Lafora Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational/genetics
7.
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
8.
Cancers (Basel) ; 12(4)2020 Apr 13.
Article in English | MEDLINE | ID: mdl-32295073

ABSTRACT

Gynecologic cancers cause over 600,000 deaths annually in women worldwide. The development of chemoresistance after initial rounds of chemotherapy contributes to tumor relapse and death due to gynecologic malignancies. In this regard, cancer stem cells (CSCs), a subpopulation of stem cells with the ability to undergo self-renewal and clonal evolution, play a key role in tumor progression and drug resistance. Aldehyde dehydrogenases (ALDH) are a group of enzymes shown to be robust CSC markers in gynecologic and other malignancies. These enzymes also play functional roles in CSCs, including detoxification of aldehydes, scavenging of reactive oxygen species (ROS), and retinoic acid (RA) signaling, making ALDH an attractive therapeutic target in various clinical scenarios. In this review, we discuss the critical roles of the ALDH in driving stemness in different gynecologic malignancies. We review inhibitors of ALDH, both general and isoform-specific, which have been used to target CSCs in gynecologic cancers. Many of these inhibitors have been shown to be effective in preclinical models of gynecologic malignancies, supporting further development in the clinic. Furthermore, ALDH inhibitors, including 673A and CM037, synergize with chemotherapy to reduce tumor growth. Thus, ALDH-targeted therapies hold promise for improving patient outcomes in gynecologic malignancies.

9.
J Med Chem ; 63(7): 3538-3551, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32134266

ABSTRACT

The overaccumulation of glycogen appears as a hallmark in various glycogen storage diseases (GSDs), including Pompe, Cori, Andersen, and Lafora disease. Accumulating evidence suggests that suppression of glycogen accumulation represents a potential therapeutic approach for treating these GSDs. Using a fluorescence polarization assay designed to screen for inhibitors of the key glycogen synthetic enzyme, glycogen synthase (GS), we identified a substituted imidazole, (rac)-2-methoxy-4-(1-(2-(1-methylpyrrolidin-2-yl)ethyl)-4-phenyl-1H-imidazol-5-yl)phenol (H23), as a first-in-class inhibitor for yeast GS 2 (yGsy2p). Data from X-ray crystallography at 2.85 Å, as well as kinetic data, revealed that H23 bound within the uridine diphosphate glucose binding pocket of yGsy2p. The high conservation of residues between human and yeast GS in direct contact with H23 informed the development of around 500 H23 analogs. These analogs produced a structure-activity relationship profile that led to the identification of a substituted pyrazole, 4-(4-(4-hydroxyphenyl)-3-(trifluoromethyl)-1H-pyrazol-5-yl)pyrogallol, with a 300-fold improved potency against human GS. These substituted pyrazoles possess a promising scaffold for drug development efforts targeting GS activity in GSDs associated with excess glycogen accumulation.


Subject(s)
Enzyme Inhibitors/chemistry , Glycogen Synthase/antagonists & inhibitors , Imidazoles/chemistry , Pyrazoles/chemistry , Animals , Caenorhabditis elegans/enzymology , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Glycogen Synthase/chemistry , Glycogen Synthase/metabolism , HEK293 Cells , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Kinetics , Molecular Structure , Protein Binding , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Saccharomyces cerevisiae/enzymology , Structure-Activity Relationship
10.
Epilepsy Behav ; 103(Pt A): 106839, 2020 02.
Article in English | MEDLINE | ID: mdl-31932179

ABSTRACT

Lafora disease (LD) is both a fatal childhood epilepsy and a glycogen storage disease caused by recessive mutations in either the Epilepsy progressive myoclonus 2A (EPM2A) or EPM2B genes. Hallmarks of LD are aberrant, cytoplasmic carbohydrate aggregates called Lafora bodies (LBs) that are a disease driver. The 5th International Lafora Epilepsy Workshop was recently held in Alcala de Henares, Spain. The workshop brought together nearly 100 clinicians, academic and industry scientists, trainees, National Institutes of Health (NIH) representation, and friends and family members of patients with LD. The workshop covered aspects of LD ranging from defining basic scientific mechanisms to elucidating a LD therapy or cure and a recently launched LD natural history study.


Subject(s)
Congresses as Topic/trends , Education/trends , Internationality , Lafora Disease/therapy , Animals , Humans , Lafora Disease/epidemiology , Lafora Disease/genetics , Mutation/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Spain/epidemiology
11.
Cell Rep ; 26(11): 3061-3075.e6, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30865894

ABSTRACT

Ovarian cancer is typified by the development of chemotherapy resistance. Chemotherapy resistance is associated with high aldehyde dehydrogenase (ALDH) enzymatic activity, increased cancer "stemness," and expression of the stem cell marker CD133. As such, ALDH activity has been proposed as a therapeutic target. Although it remains controversial which of the 19 ALDH family members drive chemotherapy resistance, ALDH1A family members have been primarily linked with chemotherapy resistant and stemness. We identified two ALDH1A family selective inhibitors (ALDH1Ai). ALDH1Ai preferentially kills CD133+ ovarian cancer stem-like cells (CSCs). ALDH1Ai induce necroptotic CSC death, mediated, in part, by the induction of mitochondrial uncoupling proteins and reduction in oxidative phosphorylation. ALDH1Ai is highly synergistic with chemotherapy, reducing tumor initiation capacity and increasing tumor eradication in vivo. These studies link ALDH1A with necroptosis and confirm the family as a critical therapeutic target to overcome chemotherapy resistance and improve patient outcomes.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Necroptosis , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Retinal Dehydrogenase/antagonists & inhibitors , AC133 Antigen/genetics , AC133 Antigen/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Mice , Neoplastic Stem Cells/drug effects , Oxidative Phosphorylation , Retinal Dehydrogenase/metabolism
12.
Cell Chem Biol ; 25(12): 1456-1469.e6, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30293938

ABSTRACT

5-Nitrofurans are antibiotic pro-drugs that have potential as cancer therapeutics. Here, we show that 5-nitrofurans can be bio-activated by aldehyde dehydrogenase (ALDH) 1A1/1A3 enzymes that are highly expressed in a subpopulation of cancer-initiating (stem) cells. We discover that the 5-nitrofuran, nifuroxazide, is selective for bio-activation by ALDH1 isoforms over ALDH2, whereby it both oxidizes ALDH1 and is converted to cytotoxic metabolites in a two-hit pro-drug mechanism. We show that ALDH1High melanoma cells are sensitive to nifuroxazide, while ALDH1A3 loss-of-function mutations confer drug resistance. In tumors, nifuroxazide targets ALDH1High melanoma subpopulations with the subsequent loss of melanoma-initiating cell potential. BRAF and MEK inhibitor therapy increases ALDH1 expression in patient melanomas, and effectively combines with nifuroxazide in melanoma cell models. The selective eradication of ALDH1High cells by nifuroxazide-ALDH1 activation goes beyond current strategies based on inhibiting ALDH1 and provides a rational basis for the nifuroxazide mechanism of action in cancer.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Hydroxybenzoates/metabolism , Hydroxybenzoates/pharmacology , Isoenzymes/metabolism , Melanoma/drug therapy , Melanoma/pathology , Neoplastic Stem Cells/drug effects , Nitrofurans/metabolism , Nitrofurans/pharmacology , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Hydroxybenzoates/chemistry , Isoenzymes/antagonists & inhibitors , Melanoma/genetics , Melanoma/metabolism , Mice , Molecular Structure , Neoplastic Stem Cells/pathology , Nitrofurans/chemistry , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacology , Retinal Dehydrogenase/antagonists & inhibitors
13.
J Med Chem ; 61(19): 8754-8773, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30221940

ABSTRACT

Aldehyde dehydrogenase (ALDH) activity is commonly used as a marker to identify cancer stem-like cells. The three ALDH1A isoforms have all been individually implicated in cancer stem-like cells and in chemoresistance; however, which isoform is preferentially expressed varies between cell lines. We sought to explore the structural determinants of ALDH1A isoform selectivity in a series of small-molecule inhibitors in support of research into the role of ALDH1A in cancer stem cells. An SAR campaign guided by a cocrystal structure of the HTS hit CM39 (7) with ALDH1A1 afforded first-in-class inhibitors of the ALDH1A subfamily with excellent selectivity over the homologous ALDH2 isoform. We also discovered the first reported modestly selective single isoform 1A2 and 1A3 inhibitors. Two compounds, 13g and 13h, depleted the CD133+ putative cancer stem cell pool, synergized with cisplatin, and achieved efficacious concentrations in vivo following IP administration. Compound 13h additionally synergized with cisplatin in a patient-derived ovarian cancer spheroid model.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/drug therapy , Aldehyde Dehydrogenase 1 Family , Animals , Cell Proliferation , Drug Discovery , Drug Screening Assays, Antitumor , Enzyme Inhibitors/classification , Female , Humans , Mice , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Retinal Dehydrogenase , Structure-Activity Relationship , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
J Med Chem ; 60(6): 2439-2455, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28219011

ABSTRACT

Aldehyde dehydrogenase 2 (ALDH2), one of 19 ALDH superfamily members, catalyzes the NAD+-dependent oxidation of aldehydes to their respective carboxylic acids. In this study, we further characterized the inhibition of four psoralen and coumarin derivatives toward ALDH2 and compared them to the ALDH2 inhibitor daidzin for selectivity against five ALDH1/2 isoenzymes. Compound 2 (Ki = 19 nM) binds within the aldehyde-binding site of the free enzyme species of ALDH2. Thirty-three structural analogs were examined to develop a stronger SAR profile. Seven compounds maintained or improved upon the selectivity toward one of the five ALDH1/2 isoenzymes, including compound 36, a selective inhibitor for ALDH2 (Ki = 2.4 µM), and compound 32, which was 10-fold selective for ALDH1A1 (Ki = 1.2 µM) versus ALDH1A2. Further medicinal chemistry on the compounds' basic scaffold could enhance the potency and selectivity for ALDH1A1 or ALDH2 and generate chemical probes to examine the unique and overlapping functions of the ALDH1/2 isoenzymes.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Aldehyde Dehydrogenase/antagonists & inhibitors , Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , Ficusin/pharmacology , Isoenzymes/antagonists & inhibitors , Retinal Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/chemistry , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Coumarins/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Ficusin/chemistry , Humans , Isoenzymes/metabolism , Molecular Docking Simulation , Retinal Dehydrogenase/chemistry , Retinal Dehydrogenase/metabolism
15.
Cell Stem Cell ; 20(3): 303-314.e5, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28041894

ABSTRACT

Lack of sensitive single-cell analysis tools has limited the characterization of metabolic activity in cancer stem cells. By hyperspectral-stimulated Raman scattering imaging of single living cells and mass spectrometry analysis of extracted lipids, we report here significantly increased levels of unsaturated lipids in ovarian cancer stem cells (CSCs) as compared to non-CSCs. Higher lipid unsaturation levels were also detected in CSC-enriched spheroids compared to monolayer cultures of ovarian cancer cell lines or primary cells. Inhibition of lipid desaturases effectively eliminated CSCs, suppressed sphere formation in vitro, and blocked tumor initiation capacity in vivo. Mechanistically, we demonstrate that nuclear factor κB (NF-κB) directly regulates the expression levels of lipid desaturases, and inhibition of desaturases blocks NF-κB signaling. Collectively, our findings reveal that increased lipid unsaturation is a metabolic marker for ovarian CSCs and a target for CSC-specific therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Lipids/chemistry , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Aldehyde Dehydrogenase/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Cell Separation , Down-Regulation/genetics , Fatty Acid Desaturases/metabolism , Female , Flow Cytometry , Humans , Mice, Nude , NF-kappa B/metabolism , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
16.
Biochemistry ; 56(1): 179-188, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27935293

ABSTRACT

Glycogen synthase (GS) is the rate limiting enzyme in the synthesis of glycogen. Eukaryotic GS is negatively regulated by covalent phosphorylation and allosterically activated by glucose-6-phosphate (G-6-P). To gain structural insights into the inhibited state of the enzyme, we solved the crystal structure of yGsy2-R589A/R592A to a resolution of 3.3 Å. The double mutant has an activity ratio similar to the phosphorylated enzyme and also retains the ability to be activated by G-6-P. When compared to the 2.88 Å structure of the wild-type G-6-P activated enzyme, the crystal structure of the low-activity mutant showed that the N-terminal domain of the inhibited state is tightly held against the dimer-related interface thereby hindering acceptor access to the catalytic cleft. On the basis of these two structural observations, we developed a reversible redox regulatory feature in yeast GS by substituting cysteine residues for two highly conserved arginine residues. When oxidized, the cysteine mutant enzyme exhibits activity levels similar to the phosphorylated enzyme but cannot be activated by G-6-P. Upon reduction, the cysteine mutant enzyme regains normal activity levels and regulatory response to G-6-P activation.


Subject(s)
Glycogen Synthase/genetics , Mutation , Saccharomyces cerevisiae/genetics , Crystallization , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Glucose-6-Phosphate/metabolism , Glucose-6-Phosphate/pharmacology , Glycogen/metabolism , Glycogen Synthase/chemistry , Glycogen Synthase/metabolism , Kinetics , Models, Molecular , Oxidation-Reduction , Phosphorylation , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Uridine Diphosphate Glucose/metabolism
17.
Arch Biochem Biophys ; 597: 21-9, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27036853

ABSTRACT

The storage polymer glycogen normally contains small amounts of covalently attached phosphate as phosphomonoesters at C2, C3 and C6 atoms of glucose residues. In the absence of the laforin phosphatase, as in the rare childhood epilepsy Lafora disease, the phosphorylation level is elevated and is associated with abnormal glycogen structure that contributes to the pathology. Laforin therefore likely functions in vivo as a glycogen phosphatase. The mechanism of glycogen phosphorylation is less well-understood. We have reported that glycogen synthase incorporates phosphate into glycogen via a rare side reaction in which glucose-phosphate rather than glucose is transferred to a growing polyglucose chain (Tagliabracci et al. (2011) Cell Metab13, 274-282). We proposed a mechanism to account for phosphorylation at C2 and possibly at C3. Our results have since been challenged (Nitschke et al. (2013) Cell Metab17, 756-767). Here we extend the evidence supporting our conclusion, validating the assay used for the detection of glycogen phosphorylation, measurement of the transfer of (32)P from [ß-(32)P]UDP-glucose to glycogen by glycogen synthase. The (32)P associated with the glycogen fraction was stable to ethanol precipitation, SDS-PAGE and gel filtration on Sephadex G50. The (32)P-signal was not affected by inclusion of excess unlabeled UDP before analysis or by treatment with a UDPase, arguing against the signal being due to contaminating [ß-(32)P]UDP generated in the reaction. Furthermore, [(32)P]UDP did not bind non-covalently to glycogen. The (32)P associated with glycogen was released by laforin treatment, suggesting that it was present as a phosphomonoester. The conclusion is that glycogen synthase can mediate the introduction of phosphate into glycogen, thereby providing a possible mechanism for C2, and perhaps C3, phosphorylation.


Subject(s)
Glycogen Synthase/chemistry , Glycogen/chemistry , Phosphates/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Glycogen/biosynthesis , Glycogen Synthase/metabolism , Humans , Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Sugars/metabolism
18.
Chem Biol Interact ; 234: 38-44, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25641190

ABSTRACT

In humans, the aldehyde dehydrogenase superfamily consists of 19 isoenzymes which mostly catalyze the NAD(P)(+)-dependent oxidation of aldehydes. Many of these isoenzymes have overlapping substrate specificities and therefore their potential physiological functions may overlap. Thus the development of new isoenzyme-selective probes would be able to better delineate the function of a single isoenzyme and its individual contribution to the metabolism of a particular substrate. This specific study was designed to find a novel modulator of ALDH2, a mitochondrial ALDH isoenzyme most well-known for its role in acetaldehyde oxidation. 53 compounds were initially identified to modulate the activity of ALDH2 by a high-throughput esterase screen from a library of 63,000 compounds. Of these initial 53 compounds, 12 were found to also modulate the oxidation of propionaldehyde by ALDH2. Single concentration measurements at 10µM compound were performed using ALDH1A1, ALDH1A2, ALDH1A3, ALDH2, ALDH1B1, ALDH3A1, ALDH4A1, and/or ALDH5A1 to determine the selectivity of these 12 compounds toward ALDH2. Four of the twelve compounds shared an aromatic lactone structure and were found to be potent inhibitors of the ALDH1/2 isoenzymes, but have no inhibitory effect on ALDH3A1, ALDH4A1 or ALDH5A1. Two of the aromatic lactones show selectivity within the ALDH1/2 class, and one appears to be selective for ALDH2 compared to all other isoenzymes tested.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Lactones/metabolism , Retinal Dehydrogenase/antagonists & inhibitors , Acetaldehyde/metabolism , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Humans , Isoenzymes/metabolism , Mitochondria/metabolism , NADP/metabolism , Oxidation-Reduction , Retinal Dehydrogenase/metabolism , Substrate Specificity
19.
J Med Chem ; 58(4): 1964-75, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25634381

ABSTRACT

Aldehyde dehydrogenases (ALDH) catalyze the irreversible oxidation of aldehydes to their corresponding carboxylic acid. Alterations in ALDH1A1 activity are associated with such diverse diseases as cancer, Parkinson's disease, obesity, and cataracts. Inhibitors of ALDH1A1 could aid in illuminating the role of this enzyme in disease processes. However, there are no commercially available selective inhibitors for ALDH1A1. Here we characterize two distinct chemical classes of inhibitors that are selective for human ALDH1A1 compared to eight other ALDH isoenzymes. The prototypical members of each structural class, CM026 and CM037, exhibit submicromolar inhibition constants but have different mechanisms of inhibition. The crystal structures of these compounds bound to ALDH1A1 demonstrate that they bind within the aldehyde binding pocket of ALDH1A1 and exploit the presence of a unique glycine residue to achieve their selectivity. These two novel and selective ALDH1A1 inhibitors may serve as chemical tools to better understand the contributions of ALDH1A1 to normal biology and to disease states.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/classification , Enzyme Inhibitors/pharmacology , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mice , Mice, Knockout , Models, Molecular , Molecular Structure , Retinal Dehydrogenase , Structure-Activity Relationship
20.
Proc Natl Acad Sci U S A ; 112(2): 458-63, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25453080

ABSTRACT

Paleogenetics is an emerging field that resurrects ancestral proteins from now-extinct organisms to test, in the laboratory, models of protein function based on natural history and Darwinian evolution. Here, we resurrect digestive alcohol dehydrogenases (ADH4) from our primate ancestors to explore the history of primate-ethanol interactions. The evolving catalytic properties of these resurrected enzymes show that our ape ancestors gained a digestive dehydrogenase enzyme capable of metabolizing ethanol near the time that they began using the forest floor, about 10 million y ago. The ADH4 enzyme in our more ancient and arboreal ancestors did not efficiently oxidize ethanol. This change suggests that exposure to dietary sources of ethanol increased in hominids during the early stages of our adaptation to a terrestrial lifestyle. Because fruit collected from the forest floor is expected to contain higher concentrations of fermenting yeast and ethanol than similar fruits hanging on trees, this transition may also be the first time our ancestors were exposed to (and adapted to) substantial amounts of dietary ethanol.


Subject(s)
Ethanol/metabolism , Hominidae/genetics , Hominidae/metabolism , Adaptation, Physiological/genetics , Alcohol Dehydrogenase/classification , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Amino Acid Sequence , Animals , Diet , Evolution, Molecular , Fermentation/genetics , Fruit/metabolism , Genetic Variation , Humans , Kinetics , Molecular Sequence Data , Phylogeny , Primates/genetics , Primates/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL