Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
J Inherit Metab Dis ; 44(1): 178-192, 2021 01.
Article in English | MEDLINE | ID: mdl-33200442

ABSTRACT

Pyridoxine-dependent epilepsy (PDE-ALDH7A1) is an autosomal recessive condition due to a deficiency of α-aminoadipic semialdehyde dehydrogenase, which is a key enzyme in lysine oxidation. PDE-ALDH7A1 is a developmental and epileptic encephalopathy that was historically and empirically treated with pharmacologic doses of pyridoxine. Despite adequate seizure control, most patients with PDE-ALDH7A1 were reported to have developmental delay and intellectual disability. To improve outcome, a lysine-restricted diet and competitive inhibition of lysine transport through the use of pharmacologic doses of arginine have been recommended as an adjunct therapy. These lysine-reduction therapies have resulted in improved biochemical parameters and cognitive development in many but not all patients. The goal of these consensus guidelines is to re-evaluate and update the two previously published recommendations for diagnosis, treatment, and follow-up of patients with PDE-ALDH7A1. Members of the International PDE Consortium initiated evidence and consensus-based process to review previous recommendations, new research findings, and relevant clinical aspects of PDE-ALDH7A1. The guideline development group included pediatric neurologists, biochemical geneticists, clinical geneticists, laboratory scientists, and metabolic dieticians representing 29 institutions from 16 countries. Consensus guidelines for the diagnosis and management of patients with PDE-ALDH7A1 are provided.


Subject(s)
Arginine/administration & dosage , Dietary Supplements , Epilepsy/diet therapy , Epilepsy/diagnosis , Aldehyde Dehydrogenase/deficiency , Consensus , Epilepsy/drug therapy , Humans , International Cooperation , Lysine/deficiency , Pyridoxine/therapeutic use
2.
J Hepatol ; 72(4): 725-735, 2020 04.
Article in English | MEDLINE | ID: mdl-31726117

ABSTRACT

BACKGROUND & AIM: Under the regulation of various oncogenic pathways, cancer cells undergo adaptive metabolic programming to maintain specific metabolic states that support their uncontrolled proliferation. As it has been difficult to directly and effectively inhibit oncogenic signaling cascades with pharmaceutical compounds, focusing on the downstream metabolic pathways that enable indefinite growth may provide therapeutic opportunities. Thus, we sought to characterize metabolic changes in hepatocellular carcinoma (HCC) development and identify metabolic targets required for tumorigenesis. METHODS: We compared gene expression profiles of Morris Hepatoma (MH3924a) and DEN (diethylnitrosamine)-induced HCC models to those of liver tissues from normal and rapidly regenerating liver models, and performed gain- and loss-of-function studies of the identified gene targets for their roles in cancer cell proliferation in vitro and in vivo. RESULTS: The proline biosynthetic enzyme PYCR1 (pyrroline-5-carboxylate reductase 1) was identified as one of the most upregulated genes in the HCC models. Knockdown of PYCR1 potently reduced cell proliferation of multiple HCC cell lines in vitro and tumor growth in vivo. Conversely, overexpression of PYCR1 enhanced the proliferation of the HCC cell lines. Importantly, PYCR1 expression was not elevated in the regenerating liver, and KD or overexpression of PYCR1 had no effect on proliferation of non-cancerous cells. Besides PYCR1, we found that additional proline biosynthetic enzymes, such as ALDH18A1, were upregulated in HCC models and also regulated HCC cell proliferation. Clinical data demonstrated that PYCR1 expression was increased in HCC, correlated with tumor grade, and was an independent predictor of clinical outcome. CONCLUSION: Enhanced expression of proline biosynthetic enzymes promotes HCC cell proliferation. Inhibition of PYCR1 or ALDH18A1 may be a novel therapeutic strategy to target HCC. LAY SUMMARY: Even with the recently approved immunotherapies against liver cancer, currently available medications show limited clinical benefits or efficacy in the majority of patients. As such, it remains a top priority to discover new targets for effective liver cancer treatment. Here, we identify a critical role for the proline biosynthetic pathway in liver cancer development, and demonstrate that targeting key proteins in the pathway, namely PYCR1 and ALDH18A1, may be a novel therapeutic strategy for liver cancer.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms/metabolism , Proline/biosynthesis , Signal Transduction/genetics , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , Diethylnitrosamine/adverse effects , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , HaCaT Cells , Hep G2 Cells , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Pyrroline Carboxylate Reductases/deficiency , Pyrroline Carboxylate Reductases/genetics , Rats , Transcriptome , Transfection , Tumor Burden/genetics , Xenograft Model Antitumor Assays , delta-1-Pyrroline-5-Carboxylate Reductase
3.
J Nutr ; 150(Suppl 1): 2556S-2560S, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33000154

ABSTRACT

Lysine is an essential amino acid, and inherited diseases of its metabolism therefore represent defects of lysine catabolism. Although some of these enzyme defects are not well described yet, glutaric aciduria type I (GA1) and antiquitin (2-aminoadipic-6-semialdehyde dehydrogenase) deficiency represent the most well-characterized diseases. GA1 is an autosomal recessive disorder due to a deficiency of glutaryl-CoA dehydrogenase. Untreated patients exhibit early onset macrocephaly and may present a neurological deterioration with regression and movement disorder at the time of a presumably "benign" infection most often during the first year of life. This is associated with a characteristic neuroimaging pattern with frontotemporal atrophy and striatal injuries. Diagnosis relies on the identification of glutaric and 3-hydroxyglutaric acid in urine along with plasma glutarylcarnitine. Treatment consists of a low-lysine diet aiming at reducing the putatively neurotoxic glutaric and 3-hydroxyglutaric acids. Additional therapeutic measures include administration of l-carnitine associated with emergency measures at the time of intercurrent illnesses aiming at preventing brain injury. Early treated (ideally through newborn screening) patients exhibit a favorable long-term neurocognitive outcome, whereas late-treated or untreated patients may present severe neurocognitive irreversible disabilities. Antiquitin deficiency is the most common form of pyridoxine-dependent epilepsy. α-Aminoadipic acid semialdehyde (AASA) and Δ-1-piperideine-6-carboxylate (P6C) accumulate proximal to the enzymatic block. P6C forms a complex with pyridoxal phosphate (PLP), a key vitamer of pyridoxine, thereby reducing PLP bioavailability and subsequently causing epilepsy. Urinary AASA is a biomarker of antiquitin deficiency. Despite seizure control, only 25% of the pyridoxine-treated patients show normal neurodevelopment. Low-lysine diet and arginine supplementation are proposed in some patients with decrease of AASA, but the impact on neurodevelopment is unclear. In summary, GA1 and antiquitin deficiency are the 2 main human defects of lysine catabolism. Both include neurological impairment. Lysine dietary restriction is a key therapy for GA1, whereas its benefits in antiquitin deficiency appear less clear.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic/metabolism , Brain/metabolism , Epilepsy/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Lysine/metabolism , 2-Aminoadipic Acid/analogs & derivatives , 2-Aminoadipic Acid/metabolism , Aldehyde Dehydrogenase/metabolism , Amino Acid Metabolism, Inborn Errors/therapy , Arginine/therapeutic use , Brain/pathology , Brain Diseases, Metabolic/therapy , Brain Diseases, Metabolic, Inborn/therapy , Carnitine/analogs & derivatives , Carnitine/metabolism , Carnitine/therapeutic use , Epilepsy/therapy , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/therapy , Pyridoxal Phosphate/metabolism , Pyridoxine/metabolism , Pyridoxine/therapeutic use
4.
J Inherit Metab Dis ; 43(4): 657-670, 2020 07.
Article in English | MEDLINE | ID: mdl-32017139

ABSTRACT

The bifunctional homooligomeric enzyme Δ1 -pyrroline-5-carboxylate synthetase (P5CS) and its encoding gene ALDH18A1 were associated with disease in 1998. Two siblings who presented paradoxical hyperammonemia (alleviated by protein), mental disability, short stature, cataracts, cutis laxa, and joint laxity, were found to carry biallelic ALDH18A1 mutations. They showed biochemical indications of decreased ornithine/proline synthesis, agreeing with the role of P5CS in the biosynthesis of these amino acids. Of 32 patients reported with this neurocutaneous syndrome, 21 familial ones hosted homozygous or compound heterozygous ALDH18A1 mutations, while 11 sporadic ones carried de novo heterozygous ALDH18A1 mutations. In 2015 to 2016, an upper motor neuron syndrome (spastic paraparesis/paraplegia SPG9) complicated with some traits of the neurocutaneous syndrome, although without report of cutis laxa, joint laxity, or herniae, was associated with monoallelic or biallelic ALDH18A1 mutations with, respectively, dominant and recessive inheritance. Of 50 SPG9 patients reported, 14 and 36 (34/2 familial/sporadic) carried, respectively, biallelic and monoallelic mutations. Thus, two neurocutaneous syndromes (recessive and dominant cutis laxa 3, abbreviated ARCL3A and ADCL3, respectively) and two SPG9 syndromes (recessive SPG9B and dominant SPG9A) are caused by essentially different spectra of ALDH18A1 mutations. On the bases of the clinical data (including our own prior patients' reports), the ALDH18A1 mutations spectra, and our knowledge on the P5CS protein, we conclude that the four syndromes share the same pathogenic mechanisms based on decreased P5CS function. Thus, these syndromes represent a continuum of increasing severity (SPG9A < SPG9B < ADCL3 ≤ ARCL3A) of the same disease, P5CS deficiency, in which the dominant mutations cause loss-of-function by dominant-negative mechanisms.


Subject(s)
Aldehyde Dehydrogenase/genetics , Bone and Bones/abnormalities , Cataract/genetics , Growth Disorders/genetics , Spastic Paraplegia, Hereditary/genetics , Aldehyde Dehydrogenase/deficiency , Humans , Mutation , Pedigree , Phenotype , Urea/metabolism
5.
J Inherit Metab Dis ; 43(4): 891-900, 2020 07.
Article in English | MEDLINE | ID: mdl-31930735

ABSTRACT

Antiquitin (ATQ) deficiency leads to tissue, plasma, and urinary accumulation of alpha-aminoadipic semialdehyde (AASA) and its Schiff base delta-1-piperideine-6-carboxylate (P6C). Although genetic testing of ALDH7A1 is the most definitive diagnostic method, quantifications of pathognomonic metabolites are important for the diagnosis and evaluation of therapeutic and dietary interventions. Current metabolite quantification methods use laborious, technically highly complex, and expensive liquid chromatography-tandem mass spectro-metry, which is available only in selected laboratories worldwide. Incubation of ortho-aminobenzaldehyde (oABA) with P6C leads to the formation of a triple aromatic ring structure with characteristic absorption and fluorescence properties. The mean concentration of P6C in nine urine samples from seven ATQ-deficient patients under standard treatment protocols was statistically highly significantly different (P < .001) compared to the mean of 74 healthy controls aged between 2 months and 57 years. Using this limited data set the specificity and sensitivity is 100% for all tested age groups using a P6C cut-off of 2.11 µmol/mmol creatinine, which represents the 99% prediction interval of the P6C concentrations in 17 control urine samples from children below 6 years of age. Plasma P6C concentrations were only elevated in one ATQ subject, possibly because P6C is trapped by pyridoxal-5-phosphate (PLP) blocking fusing with oABA. Nevertheless, both urine and plasma samples were amenable to the quantification of exogenous P6C with high response rates. The P6C quantification method using fusion of oABA with P6C is fast, simple, and inexpensive and might be readily implemented into routine clinical diagnostic laboratories for the early diagnosis of neonatal pyridoxine-dependent epilepsy.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Benzaldehydes/urine , Epilepsy/urine , Picolinic Acids/urine , Adolescent , Adult , Aldehyde Dehydrogenase/genetics , Case-Control Studies , Child , Child, Preschool , Diet , Epilepsy/diagnosis , Epilepsy/genetics , Epilepsy/metabolism , Female , Humans , Infant , Lysine/metabolism , Male , Middle Aged , Young Adult
6.
J Inherit Metab Dis ; 43(5): 1102-1111, 2020 09.
Article in English | MEDLINE | ID: mdl-32319100

ABSTRACT

Rapid diagnosis and early specific treatment of metabolic epilepsies due to inborn errors of metabolism (IEMs) is crucial to avoid irreversible sequalae. Nowadays, besides the profile analysis of amino- and organic acids, a range of additional targeted assays is used for the selective screening of those diseases. This strategy can lead to long turn-around times, repeated sampling and diagnostic delays. To replace those individual targeted assays, we developed a new liquid chromatography mass spectrometry method (LC-MS/MS) for the differential diagnosis of inherited metabolic epilepsies that are potentially treatable. The method was developed to simultaneously quantify 12 metabolites (sulfocysteine, guanidinoacetate, creatine, pipecolic acid, Δ1 -piperideine-6-carboxylate (P6C), proline, Δ1 -pyrroline-5-carboxylate (P5C), and the B6 -vitamers) enabling the diagnosis of nine different treatable IEMs presenting primarily with early-onset epilepsy. Plasma and urine samples were mixed with internal standards, precipitated and the supernatants were analyzed by LC-MS/MS. In comparison with previous assays, no derivatization of the metabolites is necessary for analysis. This LC-MS method was validated for quantitative results for all metabolites except P6C and P5C for which semiquantitative results were obtained due to the absence of commercially available standards. Coefficients of variation for all analytes were below 15% and recovery rates range between 80% and 120%. Analysis of patient samples with known IEMs demonstrated the diagnostic value of the method. The presented assay covers a selected panel of biochemical markers, improves the efficiency in the laboratory, and potentially leads to faster diagnoses and earlier treatment avoiding irreversible damage in patients affected with IEMs.


Subject(s)
Chromatography, Liquid/methods , Epilepsy/blood , Metabolism, Inborn Errors/blood , Seizures/blood , Tandem Mass Spectrometry/methods , Aldehyde Dehydrogenase/blood , Aldehyde Dehydrogenase/deficiency , Biomarkers/blood , Diagnosis, Differential , Epilepsy/diagnosis , Humans , Metabolism, Inborn Errors/diagnosis , Picolinic Acids/blood , Pipecolic Acids/blood , Seizures/diagnosis
7.
J Inherit Metab Dis ; 42(4): 620-628, 2019 07.
Article in English | MEDLINE | ID: mdl-30767241

ABSTRACT

Deficiency of antiquitin (ATQ), an enzyme involved in lysine degradation, is the major cause of vitamin B6 -dependent epilepsy. Accumulation of the potentially neurotoxic α-aminoadipic semialdehyde (AASA) may contribute to frequently associated developmental delay. AASA is formed by α-aminoadipic semialdehyde synthase (AASS) via the saccharopine pathway of lysine degradation, or, as has been postulated, by the pipecolic acid (PA) pathway, and then converted to α-aminoadipic acid by ATQ. The PA pathway has been considered to be the predominant pathway of lysine degradation in mammalian brain; however, this was refuted by recent studies in mouse. Consequently, inhibition of AASS was proposed as a potential new treatment option for ATQ deficiency. It is therefore of utmost importance to determine whether the saccharopine pathway is also predominant in human brain cells. The route of lysine degradation was analyzed by isotopic tracing studies in cultured human astrocytes, ReNcell CX human neuronal progenitor cells and human fibroblasts, and expression of enzymes of the two lysine degradation pathways was determined by Western blot. Lysine degradation was only detected through the saccharopine pathway in all cell types studied. The enrichment of 15 N-glutamate as a side product of AASA formation through AASS furthermore demonstrated activity of the saccharopine pathway. We provide first evidence that the saccharopine pathway is the major route of lysine degradation in cultured human brain cells. These results support inhibition of the saccharopine pathway as a new treatment option for ATQ deficiency.


Subject(s)
2-Aminoadipic Acid/analogs & derivatives , Aldehyde Dehydrogenase/deficiency , Epilepsy/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , 2-Aminoadipic Acid/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Epilepsy/genetics , Humans , Metabolic Networks and Pathways , Pipecolic Acids/metabolism , Vitamin B 6/therapeutic use
8.
Exp Mol Pathol ; 105(1): 63-70, 2018 08.
Article in English | MEDLINE | ID: mdl-29859945

ABSTRACT

Disease progression in alcoholic and non-alcoholic fatty liver disease shows sex-specific differences and is influenced by mechanisms linked to oxidative stress. Acetaldehyde plays a critical pathogenic role but its effects are mitigated by the activity of aldehyde dehydrogenases. Aldehyde dehydrogenase 1b1 (Aldh1b1) is the aldehyde dehydrogenase isoform with the second highest affinity for acetaldehyde after Aldh2, and is highly expressed in the intestine and liver. We examined sex differences and the effect of Aldh1b1 depletion in a murine model of chronic alcohol-induced liver disease. Male and female wild-type and Aldh1b1-depleted mice received either ethanol (10-20% v/v) in drinking water or water alone for one year, and livers were examined histopathologically, histochemically and by immunohistochemistry. A significant increase in hepatic steatosis was observed in female mice after one year of ethanol consumption, and expression of ethanol-metabolising enzymes and up-regulation by ethanol was also sex-dependent. Ethanol-induced hyperproliferation of hepatocytes was observed in female and male wild-type mice, and Aldh1b1 depletion enhanced this effect in males. Further, one ethanol-treated, Aldh1b1-depleted male developed a steatohepatitic hepatocellular carcinoma. These sex-specific differences in susceptibility to hepatic steatosis and disease progression may be related to differences in expression of ethanol-metabolising enzymes, informing the clinically significant differences. Aldh1b1 plays a role in protection from ethanol-induced hepatocellular hyperproliferation and may protect from tumour development.


Subject(s)
Alcohol Drinking/pathology , Aldehyde Dehydrogenase/deficiency , Carcinoma, Hepatocellular/pathology , Fatty Liver, Alcoholic/pathology , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/pathology , Sex Characteristics , Alcohol Drinking/adverse effects , Alcohol Drinking/metabolism , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial , Animals , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Disease Susceptibility , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Female , Liver/metabolism , Liver/pathology , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Male , Mice , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism
9.
Neuropediatrics ; 49(2): 154-157, 2018 04.
Article in English | MEDLINE | ID: mdl-29401530

ABSTRACT

Antiquitin deficiency is the most prevalent form of pyridoxine-dependent epilepsy. While most patients present with neonatal onset of therapy-resistant seizures, a few cases with late-onset during infancy have been described. Here, we describe the juvenile onset of epilepsy at the age of 17 years due to antiquitin deficiency in an Indian female with homozygosity for the most prevalent ALDH7A1 missense mutation, c.1279G > C; p.Glu427Gln in exon 14. The diagnosis was established along familial cosegregation analysis for an affected offspring, that had neonatal pyridoxine responsive seizures and had been found to be compound heterozygous for c.1279G > C; p.Glu427Gln in exon 14 and a nonsense mutation c.796C > T; p.Arg266* in exon 9. While seizures in the mother had been incompletely controlled by levetiracetam, she remained seizure-free on pyridoxine monotherapy, 200 mg/day. Her fourth pregnancy resulted in a female affected offspring, who was treated prospectively and never developed seizures with a normal outcome at age 2 years while on pyridoxine. This report illustrates that the phenotypic spectrum of antiquitin deficiency is still underestimated and that this treatable inborn error of metabolism has to be considered in case of therapy-resistant seizures even at older age. It furthermore supports prospective in utero treatment with pyridoxine in forthcoming pregnancies at risk.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Epilepsy/etiology , Epilepsy/genetics , Metabolic Diseases/complications , Metabolic Diseases/genetics , Age of Onset , Aldehyde Dehydrogenase/genetics , Epilepsy/blood , Epilepsy/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Metabolic Diseases/blood , Metabolic Diseases/diagnostic imaging , Pipecolic Acids/blood , Young Adult
10.
Nature ; 489(7417): 571-5, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22922648

ABSTRACT

Haematopoietic stem cells (HSCs) regenerate blood cells throughout the lifespan of an organism. With age, the functional quality of HSCs declines, partly owing to the accumulation of damaged DNA. However, the factors that damage DNA and the protective mechanisms that operate in these cells are poorly understood. We have recently shown that the Fanconi anaemia DNA-repair pathway counteracts the genotoxic effects of reactive aldehydes. Mice with combined inactivation of aldehyde catabolism (through Aldh2 knockout) and the Fanconi anaemia DNA-repair pathway (Fancd2 knockout) display developmental defects, a predisposition to leukaemia, and are susceptible to the toxic effects of ethanol-an exogenous source of acetaldehyde. Here we report that aged Aldh2(-/-) Fancd2(-/-) mutant mice that do not develop leukaemia spontaneously develop aplastic anaemia, with the concomitant accumulation of damaged DNA within the haematopoietic stem and progenitor cell (HSPC) pool. Unexpectedly, we find that only HSPCs, and not more mature blood precursors, require Aldh2 for protection against acetaldehyde toxicity. Additionally, the aldehyde-oxidizing activity of HSPCs, as measured by Aldefluor stain, is due to Aldh2 and correlates with this protection. Finally, there is more than a 600-fold reduction in the HSC pool of mice deficient in both Fanconi anaemia pathway-mediated DNA repair and acetaldehyde detoxification. Therefore, the emergence of bone marrow failure in Fanconi anaemia is probably due to aldehyde-mediated genotoxicity restricted to the HSPC pool. These findings identify a new link between endogenous reactive metabolites and DNA damage in HSCs, and define the protective mechanisms that counteract this threat.


Subject(s)
Aldehydes/toxicity , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mutagens/toxicity , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aging , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Animals , Bone Marrow/pathology , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair , Ethanol/toxicity , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Kaplan-Meier Estimate , Leukemia/metabolism , Leukemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Haematologica ; 102(6): 1054-1065, 2017 06.
Article in English | MEDLINE | ID: mdl-28280079

ABSTRACT

Aldehyde dehydrogenase 1A1 (ALDH1A1) activity is high in hematopoietic stem cells and functions in part to protect stem cells from reactive aldehydes and other toxic compounds. In contrast, we found that approximately 25% of all acute myeloid leukemias expressed low or undetectable levels of ALDH1A1 and that this ALDH1A1- subset of leukemias correlates with good prognosis cytogenetics. ALDH1A1- cell lines as well as primary leukemia cells were found to be sensitive to treatment with compounds that directly and indirectly generate toxic ALDH substrates including 4-hydroxynonenal and the clinically relevant compounds arsenic trioxide and 4-hydroperoxycyclophosphamide. In contrast, normal hematopoietic stem cells were relatively resistant to these compounds. Using a murine xenotransplant model to emulate a clinical treatment strategy, established ALDH1A1- leukemias were also sensitive to in vivo treatment with cyclophosphamide combined with arsenic trioxide. These results demonstrate that targeting ALDH1A1- leukemic cells with toxic ALDH1A1 substrates such as arsenic and cyclophosphamide may be a novel targeted therapeutic strategy for this subset of acute myeloid leukemias.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Drug Therapy, Combination/methods , Leukemia, Myeloid, Acute/drug therapy , Aldehyde Dehydrogenase 1 Family , Animals , Arsenic Trioxide , Arsenicals/therapeutic use , Cells, Cultured , Cyclophosphamide/therapeutic use , Heterografts , Humans , Leukemia, Myeloid, Acute/enzymology , Mice , Molecular Targeted Therapy , Oxides/therapeutic use , Retinal Dehydrogenase
12.
Nature ; 475(7354): 53-8, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734703

ABSTRACT

Reactive aldehydes are common carcinogens. They are also by-products of several metabolic pathways and, without enzymatic catabolism, may accumulate and cause DNA damage. Ethanol, which is metabolised to acetaldehyde, is both carcinogenic and teratogenic in humans. Here we find that the Fanconi anaemia DNA repair pathway counteracts acetaldehyde-induced genotoxicity in mice. Our results show that the acetaldehyde-catabolising enzyme Aldh2 is essential for the development of Fancd2(-/-) embryos. Nevertheless, acetaldehyde-catabolism-competent mothers (Aldh2(+/-)) can support the development of double-mutant (Aldh2(-/-)Fancd2(-/-)) mice. However, these embryos are unusually sensitive to ethanol exposure in utero, and ethanol consumption by postnatal double-deficient mice rapidly precipitates bone marrow failure. Lastly, Aldh2(-/-)Fancd2(-/-) mice spontaneously develop acute leukaemia. Acetaldehyde-mediated DNA damage may critically contribute to the genesis of fetal alcohol syndrome in fetuses, as well as to abnormal development, haematopoietic failure and cancer predisposition in Fanconi anaemia patients.


Subject(s)
Aldehydes/antagonists & inhibitors , Aldehydes/toxicity , Fanconi Anemia Complementation Group D2 Protein/metabolism , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Alleles , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/physiopathology , Cell Line , Cell Survival/drug effects , Chickens , Clone Cells/drug effects , DNA Damage/genetics , DNA Repair/genetics , Embryo Loss/chemically induced , Embryo Loss/etiology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Ethanol/metabolism , Ethanol/toxicity , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Fetal Alcohol Spectrum Disorders/etiology , Gene Deletion , Genes, Essential , Hematopoiesis/drug effects , Male , Mice , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Pregnancy , Teratogens/metabolism , Teratogens/toxicity , Weaning
13.
Biochim Biophys Acta ; 1852(2): 310-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25086229

ABSTRACT

Mitochondrial aldehyde dehydrogenase 2 (ALDH2) was demonstrated to play cardioprotective roles in cardiovascular diseases. Nonetheless, little is known about the roles and mechanisms of ALDH2 in pressure overload-induced cardiac damages. In this study, we revealed that ALDH2 deficiency overtly exacerbated transverse aortic constriction (TAC)-induced cardiac dysfunction. Cardiomyocyte enlargement was observed in both WT and ALDH2-/- mice in HE-stained myocardial tissue samples at 8 weeks post TAC surgery. Mitochondrial morphology and structure were also significantly damaged post TAC surgery and the changes were aggravated in ALDH2-/- TAC hearts. ALDH2 deficiency also depressed myocardial autophagy in hearts at 8 weeks post TAC surgery with a potential mechanism of repressing the expression of Beclin-1 and promoting the interaction between Bcl-2 and Beclin-1. These data indicate that ALDH2 deficiency exacerbates the pressure overload induced cardiac dysfunction partly by inhibiting Beclin-1 dependent autophagy pathway. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Apoptosis Regulatory Proteins/metabolism , Autophagy , Heart/physiopathology , Signal Transduction , Adenylate Kinase/metabolism , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Animals , Beclin-1 , Blotting, Western , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Male , Mice, Inbred C57BL , Models, Biological , Myocardium/pathology , Myocardium/ultrastructure , Phosphorylation , Pressure , TOR Serine-Threonine Kinases/metabolism , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
14.
Circulation ; 131(19): 1665-73, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25759460

ABSTRACT

BACKGROUND: Coronary spastic angina (CSA) is a common disease among East Asians, including Japanese. The prevalence of alcohol flushing syndrome associated with deficient activity of the variant aldehyde dehydrogenase 2 (ALDH2*2) genotype is prevalent among East Asians. We examined whether CSA is associated with the ALDH2*2 genotype in Japanese. METHODS AND RESULTS: The study subjects consisted of 202 patients in whom intracoronary injection of acetylcholine was performed by angiography on suspicion of CSA (119 men and 83 women; mean age, 66.2±11.4 years). They were divided into CSA (112 patients) and control groups (90 patients). ALDH2 genotyping was performed by the direct application of the TaqMan polymerase chain reaction system on dried whole blood. Clinical and laboratory data were examined using conventional methods. The frequencies of male sex, ALDH2*2 genotype carriers, alcohol flushing syndrome, tobacco smoking, and the plasma level of uric acid were higher (P<0.001, P<0.001, P<0.001, P<0.001, and P=0.007, respectively) and the plasma high-density lipoprotein cholesterol levels were lower (P<0.001) in the CSA group than in the control group. The multivariable logistic regression analysis revealed that ALDH2*2 genotype and smoking were significantly associated with CSA (P<0.001 and P=0.024, respectively). CONCLUSIONS: East Asian variant ALDH2*2 genotypes and, hence, deficient ALDH2 activity were associated with CSA in Japanese. These data support further investigation of treatment targeting aldehydes for CSA.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Aldehydes/metabolism , Coronary Vasospasm/genetics , Ethanol/adverse effects , Flushing/chemically induced , Acetylcholine , Aged , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Cholesterol, HDL/blood , Coronary Angiography , Coronary Vasospasm/diagnostic imaging , Coronary Vasospasm/enzymology , Coronary Vasospasm/ethnology , Coronary Vessels , Female , Genotype , Humans , Injections, Intra-Arterial , Japan , Lipid Peroxidation , Male , Middle Aged , Oxidative Stress , Polymorphism, Single Nucleotide , Risk Factors , Smoking/epidemiology , Uric Acid/blood
15.
Hepatology ; 60(1): 146-57, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24492981

ABSTRACT

UNLABELLED: Aldehyde dehydrogenase 2 (ALDH2) is the major enzyme that metabolizes acetaldehyde produced from alcohol metabolism. Approximately 40-50% of East Asians carry an inactive ALDH2 gene and exhibit acetaldehyde accumulation after alcohol consumption. However, the role of ALDH2 deficiency in the pathogenesis of alcoholic liver injury remains obscure. In the present study, wild-type and ALDH2(-/-) mice were subjected to ethanol feeding and/or carbon tetrachloride (CCl4 ) treatment, and liver injury was assessed. Compared with wild-type mice, ethanol-fed ALDH2(-/-) mice had higher levels of malondialdehyde-acetaldehyde (MAA) adduct and greater hepatic inflammation, with higher hepatic interleukin (IL)-6 expression but surprisingly lower levels of steatosis and serum alanine aminotransferase (ALT). Higher IL-6 levels were also detected in ethanol-treated precision-cut liver slices from ALDH2(-/-) mice and in Kupffer cells isolated from ethanol-fed ALDH2(-/-) mice than those levels in wild-type mice. In vitro incubation with MAA enhanced the lipopolysaccharide (LPS)-mediated stimulation of IL-6 production in Kupffer cells. In agreement with these findings, hepatic activation of the major IL-6 downstream signaling molecule signal transducer and activator of transcription 3 (STAT3) was higher in ethanol-fed ALDH2(-/-) mice than in wild-type mice. An additional deletion of hepatic STAT3 increased steatosis and hepatocellular damage in ALDH2(-/-) mice. Finally, ethanol-fed ALDH2(-/-) mice were more prone to CCl4 -induced liver inflammation and fibrosis than ethanol-fed wild-type mice. CONCLUSION: ALDH2(-/-) mice are resistant to ethanol-induced steatosis but prone to inflammation and fibrosis by way of MAA-mediated paracrine activation of IL-6 in Kupffer cells. These findings suggest that alcohol, by way of acetaldehyde and its associated adducts, stimulates hepatic inflammation and fibrosis independent from causing hepatocyte death, and that ALDH2-deficient individuals may be resistant to steatosis and blood ALT elevation, but are prone to liver inflammation and fibrosis following alcohol consumption.


Subject(s)
Aldehyde Dehydrogenase/genetics , Fatty Liver, Alcoholic/enzymology , Hepatitis/enzymology , Liver Cirrhosis/enzymology , Acetaldehyde/metabolism , Aldehyde Dehydrogenase/blood , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial , Animals , Carbon Tetrachloride Poisoning/enzymology , Carbon Tetrachloride Poisoning/genetics , Central Nervous System Depressants/pharmacokinetics , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Cytochrome P-450 CYP2E1/metabolism , Ethanol/pharmacokinetics , Fatty Liver, Alcoholic/genetics , Female , Hepatitis/genetics , Isoenzymes/metabolism , Kupffer Cells/enzymology , Liver Cirrhosis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/physiology , Retinal Dehydrogenase/metabolism , STAT3 Transcription Factor/metabolism
16.
Alcohol Clin Exp Res ; 39(8): 1465-75, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26173414

ABSTRACT

BACKGROUND: Acetaldehyde, the toxic ethanol (EtOH) metabolite, disrupts intestinal epithelial barrier function. Aldehyde dehydrogenase (ALDH) detoxifies acetaldehyde into acetate. Subpopulations of Asians and Native Americans show polymorphism with loss-of-function mutations in ALDH2. We evaluated the effect of ALDH2 deficiency on EtOH-induced disruption of intestinal epithelial tight junctions and adherens junctions, gut barrier dysfunction, and liver injury. METHODS: Wild-type and ALDH2-deficient mice were fed EtOH (1 to 6%) in Lieber-DeCarli diet for 4 weeks. Gut permeability in vivo was measured by plasma-to-luminal flux of FITC-inulin, tight junction and adherens junction integrity was analyzed by confocal microscopy, and liver injury was assessed by the analysis of plasma transaminase activity, histopathology, and liver triglyceride. RESULTS: EtOH feeding elevated colonic mucosal acetaldehyde, which was significantly greater in ALDH2-deficient mice. ALDH2(-/-) mice showed a drastic reduction in the EtOH diet intake. Therefore, this study was continued only in wild-type and ALDH2(+/-) mice. EtOH feeding elevated mucosal inulin permeability in distal colon, but not in proximal colon, ileum, or jejunum of wild-type mice. In ALDH2(+/-) mice, EtOH-induced inulin permeability in distal colon was not only higher than that in wild-type mice, but inulin permeability was also elevated in the proximal colon, ileum, and jejunum. Greater inulin permeability in distal colon of ALDH2(+/-) mice was associated with a more severe redistribution of tight junction and adherens junction proteins from the intercellular junctions. In ALDH2(+/-) mice, but not in wild-type mice, EtOH feeding caused a loss of junctional distribution of tight junction and adherens junction proteins in the ileum. Histopathology, plasma transaminases, and liver triglyceride analyses showed that EtOH-induced liver damage was significantly greater in ALDH2(+/-) mice compared to wild-type mice. CONCLUSIONS: These data demonstrate that ALDH2 deficiency enhances EtOH-induced disruption of intestinal epithelial tight junctions, barrier dysfunction, and liver damage.


Subject(s)
Aldehyde Dehydrogenase/deficiency , Ethanol/toxicity , Fatty Liver/chemically induced , Fatty Liver/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Animals , Fatty Liver/pathology , Female , Gastrointestinal Absorption/drug effects , Gastrointestinal Absorption/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tight Junctions/pathology
17.
Arterioscler Thromb Vasc Biol ; 34(4): 894-901, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24558108

ABSTRACT

OBJECTIVE: Mesenchymal stem cell (MSC) therapy is a promising treatment for ischemic injury. However, the environmental regulatory mechanism is essentially unclear and thus greatly limits its application in clinical setting. Accumulating evidence suggests a vital role of aldehyde dehydrogenase-2 (ALDH2) in microenvironment homeostasis after ischemia. About 540 million people or 8% of population worldwide carry a loss-of-function allele of ALDH2. It is unknown whether ALDH2 functions as a host factor regulating the therapeutic potential of donor MSCs. Therefore, this study was designed to determine whether and how host ALDH2 regulates MSC retention and therapeutic efficacy after transplantation into ischemic tissues. APPROACH AND RESULTS: Mice limb ischemia was performed by femoral artery ligation. A total of 10(6) MSCs were injected into the ischemic thigh muscles. One, 2, and 4 weeks after transplantation, MSC retention, blood perfusion recovery, limb necrosis, and fibrosis were analyzed. Compared with wild-type tissue, ALDH2 deficiency tissue significantly limited MSC retention and its perfusion recovery and limb salvage effects after ischemia. Importantly, local overexpression of ALDH2 optimized tissue microenvironment and significantly magnified all these MSC-induced improvement. Further study indicated that host ALDH2 regulated transplanted MSC survival and therapy as a microenvironment homeostasis mediator via local capillary density, energy supply, and oxidative stress regulating after ischemia. CONCLUSIONS: Our study establishes ALDH2 as a key mediator of host stem cell niche for optimal MSC therapy and suggests that ALDH2 deficiency present in the general population is a limiting host factor to be considered for MSC therapy.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Ischemia/surgery , Mesenchymal Stem Cell Transplantation , Muscle, Skeletal/blood supply , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Animals , Cells, Cultured , Disease Models, Animal , Energy Metabolism , Hindlimb , Ischemia/enzymology , Ischemia/genetics , Ischemia/physiopathology , Limb Salvage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Oxidative Stress , Recovery of Function , Regional Blood Flow , Stem Cell Niche , Time Factors , Transfection
18.
Adv Exp Med Biol ; 815: 71-88, 2015.
Article in English | MEDLINE | ID: mdl-25427902

ABSTRACT

Among various potential mechanisms that could explain alcohol carcinogenicity, the metabolism of ethanol to acetaldehyde represents an obvious possible mechanism, at least in some tissues. The fundamental principle of genotoxic carcinogenesis is the formation of mutagenic DNA adducts in proliferating cells. If not repaired, these adducts can result in mutations during DNA replication, which are passed on to cells during mitosis. Consistent with a genotoxic mechanism, acetaldehyde does react with DNA to form a variety of different types of DNA adducts. In this chapter we will focus more specifically on N2-ethylidene-deoxyguanosine (N2-ethylidene-dG), the major DNA adduct formed from the reaction of acetaldehyde with DNA and specifically highlight recent data on the measurement of this DNA adduct in the human body after alcohol exposure. Because results are of particular biological relevance for alcohol-related cancer of the upper aerodigestive tract (UADT), we will also discuss the histology and cytology of the UADT, with the goal of placing the adduct data in the relevant cellular context for mechanistic interpretation. Furthermore, we will discuss the sources and concentrations of acetaldehyde and ethanol in different cell types during alcohol consumption in humans. Finally, in the last part of the chapter, we will critically evaluate the concept of carcinogenic levels of acetaldehyde, which has been raised in the literature, and discuss how data from acetaldehyde genotoxicity are and can be utilized in physiologically based models to evaluate exposure risk.


Subject(s)
Acetaldehyde/metabolism , DNA Adducts/toxicity , Ethanol/toxicity , Neoplasms/chemically induced , Acetaldehyde/toxicity , Alcohol Drinking/adverse effects , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase, Mitochondrial , DNA Damage , Humans
19.
J Mol Cell Cardiol ; 71: 92-104, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24434637

ABSTRACT

Mitochondrial aldehyde dehydrogenase (ALDH2) protects against cardiac injury via reducing production of 4-hydroxynonenal (4-HNE) and ROS. This study was designed to examine the impact of ALDH2 on doxorubicin (DOX)-induced cardiomyopathy and mechanisms involved with a focus on autophagy. 4-HNE and autophagic markers were detected by Western blotting in ventricular tissues from normal donors and patients with idiopathic dilated cardiomyopathy. Cardiac function, 4-HNE and levels of autophagic markers were detected in WT, ALDH2 knockout or ALDH2 transfected mice treated with or without DOX. Autophagy regulatory signaling including PI-3K, AMPK and Akt was examined in DOX-treated cardiomyocytes incubated with or without ALDH2 activator Alda-1. DOX-induced myocardial dysfunction, upregulation of 4-HNE and autophagic proteins were further aggravated in ALDH2 knockout mice while they were ameliorated in ALDH2 transfected mice. DOX downregulated Class I and upregulated Class III PI3-kinase, the effect of which was augmented by ALDH2 deletion. Accumulation of 4-HNE and autophagic protein markers in DOX-induced cardiomyocytes was significantly reduced by Alda-1. DOX depressed phosphorylated Akt but not AMPK, the effect was augmented by ALDH2 knockout. The autophagy inhibitor 3-MA attenuated, whereas autophagy inducer rapamycin mimicked DOX-induced cardiomyocyte contractile defects. In addition, rapamycin effectively mitigated Alda-1-offered protective action against DOX-induced cardiomyocyte dysfunction. Our data further revealed downregulated ALDH2 and upregulated autophagy levels in the hearts from patients with dilated cardiomyopathy. Taken together, our findings suggest that inhibition of 4-HNE and autophagy may be a plausible mechanism underscoring ALDH2-offered protection against DOX-induced cardiac defect. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".


Subject(s)
Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Cardiomyopathies/chemically induced , Cardiomyopathies/enzymology , Doxorubicin/pharmacology , Myocardial Contraction/drug effects , Myocardium/metabolism , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial , Animals , Autophagy/drug effects , Cardiomyopathies/metabolism , Down-Regulation/drug effects , Female , Heart/drug effects , Heart/physiopathology , Humans , Inactivation, Metabolic , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects
20.
Biochim Biophys Acta ; 1833(12): 3218-3227, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24080087

ABSTRACT

The deregulation of B cell differentiation has been shown to contribute to autoimmune disorders, hematological cancers, and aging. We provide evidence that the retinoic acid-producing enzyme aldehyde dehydrogenase 1a1 (Aldh1a1) is an oncogene suppressor in specific splenic IgG1(+)/CD19(-) and IgG1(+)/CD19(+) B cell populations. Aldh1a1 regulated transcription factors during B cell differentiation in a sequential manner: 1) retinoic acid receptor alpha (Rara) in IgG1(+)/CD19(-) and 2) zinc finger protein Zfp423 and peroxisome proliferator-activated receptor gamma (Pparg) in IgG1(+)/CD19(+) splenocytes. In Aldh1a1(-/-) mice, splenic IgG1(+)/CD19(-) and IgG1(+)/CD19(+) B cells acquired expression of proto-oncogenic genes c-Fos, c-Jun, and Hoxa10 that resulted in splenomegaly. Human multiple myeloma B cell lines also lack Aldh1a1 expression; however, ectopic Aldh1a1 expression rescued Rara and Znf423 expressions in these cells. Our data highlight a mechanism by which an enzyme involved in vitamin A metabolism can improve B cell resistance to oncogenesis.


Subject(s)
Aldehyde Dehydrogenase/metabolism , B-Lymphocytes/metabolism , Genes, Tumor Suppressor , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD19/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Models, Biological , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , PPAR gamma/metabolism , Response Elements/genetics , Retinal Dehydrogenase , Spleen/drug effects , Spleen/pathology , Splenomegaly/immunology , Splenomegaly/pathology , Transcription Factors/metabolism , Tretinoin/pharmacology , Vitamin A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL