Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Calcif Tissue Int ; 108(2): 265-276, 2021 02.
Article in English | MEDLINE | ID: mdl-33068139

ABSTRACT

ALDH2 inactivating mutation (ALDH2*2) is the most abundant mutation leading to bone morphological aberration. Osteoporosis has long been associated with changes in bone biomaterial in elderly populations. Such changes can be exacerbated with elevated ethanol consumption and in subjects with impaired ethanol metabolism, such as carriers of aldehyde dehydrogenase 2 (ALDH2)-deficient gene, ALDH2*2. So far, little is known about bone compositional changes besides a decrease in mineralization. Raman spectroscopic imaging has been utilized to study the changes in overall composition of C57BL/6 female femur bone sections, as well as in compound spatial distribution. Raman maps of bone sections were analyzed using multilinear regression with these four isolated components, resulting in maps of their relative distribution. A 15-week treatment of both wild-type (WT) and ALDH2*2/*2 mice with 20% ethanol in the drinking water resulted in a significantly lower mineral content (p < 0.05) in the bones. There was no significant change in mineral and collagen content due to the mutation alone (p > 0.4). Highly localized islets of elongated adipose tissue were observed on most maps. Elevated fat content was found in ALDH2*2 knock-in mice consuming ethanol (p < 0.0001) and this effect appeared cumulative. This work conclusively demonstrates that that osteocytes in femurs of older female mice accumulate fat, as has been previously theorized, and that fat accumulation is likely modulated by levels of acetaldehyde, the ethanol metabolite.


Subject(s)
Alcohol Drinking/adverse effects , Aldehyde Dehydrogenase, Mitochondrial/genetics , Cortical Bone , Ethanol , Femur , Acetaldehyde , Animals , Ethanol/administration & dosage , Female , Mice , Mice, Inbred C57BL
2.
Acta Neuropathol Commun ; 7(1): 190, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31829281

ABSTRACT

Aldehyde dehydrogenase 2 deficiency (ALDH2*2) causes facial flushing in response to alcohol consumption in approximately 560 million East Asians. Recent meta-analysis demonstrated the potential link between ALDH2*2 mutation and Alzheimer's Disease (AD). Other studies have linked chronic alcohol consumption as a risk factor for AD. In the present study, we show that fibroblasts of an AD patient that also has an ALDH2*2 mutation or overexpression of ALDH2*2 in fibroblasts derived from AD patients harboring ApoE ε4 allele exhibited increased aldehydic load, oxidative stress, and increased mitochondrial dysfunction relative to healthy subjects and exposure to ethanol exacerbated these dysfunctions. In an in vivo model, daily exposure of WT mice to ethanol for 11 weeks resulted in mitochondrial dysfunction, oxidative stress and increased aldehyde levels in their brains and these pathologies were greater in ALDH2*2/*2 (homozygous) mice. Following chronic ethanol exposure, the levels of the AD-associated protein, amyloid-ß, and neuroinflammation were higher in the brains of the ALDH2*2/*2 mice relative to WT. Cultured primary cortical neurons of ALDH2*2/*2 mice showed increased sensitivity to ethanol and there was a greater activation of their primary astrocytes relative to the responses of neurons or astrocytes from the WT mice. Importantly, an activator of ALDH2 and ALDH2*2, Alda-1, blunted the ethanol-induced increases in Aß, and the neuroinflammation in vitro and in vivo. These data indicate that impairment in the metabolism of aldehydes, and specifically ethanol-derived acetaldehyde, is a contributor to AD associated pathology and highlights the likely risk of alcohol consumption in the general population and especially in East Asians that carry ALDH2*2 mutation.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Ethanol/toxicity , Aged , Aged, 80 and over , Aldehydes , Animals , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/genetics , Ethanol/administration & dosage , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Knock-In Techniques , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Mutation/drug effects , Mutation/genetics
3.
Proc Natl Acad Sci U S A ; 115(24): 6279-6284, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29794221

ABSTRACT

Xerostomia (dry mouth) is the most common side effect of radiation therapy in patients with head and neck cancer and causes difficulty speaking and swallowing. Since aldehyde dehydrogenase 3A1 (ALDH3A1) is highly expressed in mouse salivary stem/progenitor cells (SSPCs), we sought to determine the role of ALDH3A1 in SSPCs using genetic loss-of-function and pharmacologic gain-of-function studies. Using DarkZone dye to measure intracellular aldehydes, we observed higher aldehyde accumulation in irradiated Aldh3a1-/- adult murine salisphere cells and in situ in whole murine embryonic salivary glands enriched in SSPCs compared with wild-type glands. To identify a safe ALDH3A1 activator for potential clinical testing, we screened a traditional Chinese medicine library and isolated d-limonene, commonly used as a food-flavoring agent, as a single constituent activator. ALDH3A1 activation by d-limonene significantly reduced aldehyde accumulation in SSPCs and whole embryonic glands, increased sphere-forming ability, decreased apoptosis, and improved submandibular gland structure and function in vivo after radiation. A phase 0 study in patients with salivary gland tumors showed effective delivery of d-limonene into human salivary glands following daily oral dosing. Given its safety and bioavailability, d-limonene may be a good clinical candidate for mitigating xerostomia in patients with head and neck cancer receiving radiation therapy.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Cyclohexenes/pharmacology , Radiotherapy/adverse effects , Salivary Glands/metabolism , Terpenes/pharmacology , Xerostomia/metabolism , Animals , Apoptosis/drug effects , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Limonene , Medicine, Chinese Traditional/methods , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Salivary Glands/drug effects , Salivary Glands/radiation effects , Stem Cells/drug effects , Stem Cells/metabolism , Submandibular Gland/drug effects , Submandibular Gland/metabolism , Xerostomia/drug therapy
4.
Mol Genet Metab ; 119(1-2): 28-36, 2016 09.
Article in English | MEDLINE | ID: mdl-27650066

ABSTRACT

Maintenance of the hematopoietic stem cell (HSC) compartment depends on the ability to metabolize exogenously and endogenously generated toxins, and to repair cellular damage caused by such toxins. Reactive aldehydes have been demonstrated to cause specific genotoxic injury, namely DNA interstrand cross-links. Aldehyde dehydrogenase 2 (ALDH2) is a member of a 19 isoenzyme ALDH family with different substrate specificities, subcellular localization, and patterns of expression. ALDH2 is localized in mitochondria and is essential for the metabolism of acetaldehyde, thereby placing it directly downstream of ethanol metabolism. Deficiency in ALDH2 expression and function are caused by a single nucleotide substitution and resulting amino acid change, called ALDH2*2. This genetic polymorphism affects 35-45% of East Asians (about ~560 million people), and causes the well-known Asian flushing syndrome, which results in disulfiram-like reactions after ethanol consumption. Recently, the ALDH2*2 genotype has been found to be associated with marrow failure, with both an increased risk of sporadic aplastic anemia and more rapid progression of Fanconi anemia. This review discusses the unexpected interrelationship between aldehydes, ALDH2 and hematopoietic stem cell biology, and in particular its relationship to Fanconi anemia.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/genetics , Anemia, Aplastic/genetics , Fanconi Anemia/genetics , Hematopoietic Stem Cells/enzymology , Aldehydes/metabolism , Anemia, Aplastic/enzymology , Anemia, Aplastic/pathology , Fanconi Anemia/enzymology , Fanconi Anemia/pathology , Flushing/genetics , Flushing/pathology , Humans , Polymorphism, Single Nucleotide , Substrate Specificity
5.
J Biol Chem ; 291(26): 13715-29, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129239

ABSTRACT

Inflammatory breast cancer (IBC) is an extremely lethal cancer that rapidly metastasizes. Although the molecular attributes of IBC have been described, little is known about the underlying metabolic features of the disease. Using a variety of metabolic assays, including (13)C tracer experiments, we found that SUM149 cells, the primary in vitro model of IBC, exhibit metabolic abnormalities that distinguish them from other breast cancer cells, including elevated levels of N-acetylaspartate, a metabolite primarily associated with neuronal disorders and gliomas. Here we provide the first evidence of N-acetylaspartate in breast cancer. We also report that the oncogene RhoC, a driver of metastatic potential, modulates glutamine and N-acetylaspartate metabolism in IBC cells in vitro, revealing a novel role for RhoC as a regulator of tumor cell metabolism that extends beyond its well known role in cytoskeletal rearrangement.


Subject(s)
Aspartic Acid/analogs & derivatives , Glutamine/metabolism , Inflammatory Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Aspartic Acid/biosynthesis , Aspartic Acid/genetics , Cell Line, Tumor , Female , Glutamine/genetics , Humans , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/pathology , Neoplasm Proteins/genetics , rho GTP-Binding Proteins/genetics , rhoC GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...