Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 587(7832): 98-102, 2020 11.
Article in English | MEDLINE | ID: mdl-33116305

ABSTRACT

Adipose tissue is usually classified on the basis of its function as white, brown or beige (brite)1. It is an important regulator of systemic metabolism, as shown by the fact that dysfunctional adipose tissue in obesity leads to a variety of secondary metabolic complications2,3. In addition, adipose tissue functions as a signalling hub that regulates systemic metabolism through paracrine and endocrine signals4. Here we use single-nucleus RNA-sequencing (snRNA-seq) analysis in mice and humans to characterize adipocyte heterogeneity. We identify a rare subpopulation of adipocytes in mice that increases in abundance at higher temperatures, and we show that this subpopulation regulates the activity of neighbouring adipocytes through acetate-mediated modulation of their thermogenic capacity. Human adipose tissue contains higher numbers of cells of this subpopulation, which could explain the lower thermogenic activity of human compared to mouse adipose tissue and suggests that targeting this pathway could be used to restore thermogenic activity.


Subject(s)
Adipocytes/metabolism , Cell Nucleus/genetics , RNA-Seq , Single-Cell Analysis , Thermogenesis/genetics , Acetates/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adult , Aged , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Cell Separation , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Energy Metabolism , Female , Humans , Male , Mice , Middle Aged , Paracrine Communication , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Young Adult
2.
EMBO J ; 40(20): e107680, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34532864

ABSTRACT

Cell plasticity is a crucial hallmark leading to cancer metastasis. Upregulation of Rho/ROCK pathway drives actomyosin contractility, protrusive forces, and contributes to the occurrence of highly invasive amoeboid cells in tumors. Cancer stem cells are similarly associated with metastasis, but how these populations arise in tumors is not fully understood. Here, we show that the novel oncogene RASSF1C drives mesenchymal-to-amoeboid transition and stem cell attributes in breast cancer cells. Mechanistically, RASSF1C activates Rho/ROCK via SRC-mediated RhoGDI inhibition, resulting in generation of actomyosin contractility. Moreover, we demonstrate that RASSF1C-induced amoeboid cells display increased expression of cancer stem-like markers such as CD133, ALDH1, and Nanog, and are accompanied by higher invasive potential in vitro and in vivo. Further, RASSF1C-induced amoeboid cells employ extracellular vesicles to transfer the invasive phenotype to target cells and tissue. Importantly, the underlying RASSF1C-driven biological processes concur to explain clinical data: namely, methylation of the RASSF1C promoter correlates with better survival in early-stage breast cancer patients. Therefore, we propose the use of RASSF1 gene promoter methylation status as a biomarker for patient stratification.


Subject(s)
Breast Neoplasms/genetics , Extracellular Vesicles/metabolism , Neoplastic Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , rhoA GTP-Binding Protein/genetics , src-Family Kinases/genetics , AC133 Antigen/genetics , AC133 Antigen/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , CpG Islands , DNA Methylation , Extracellular Vesicles/chemistry , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mice, SCID , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
3.
Exp Cell Res ; 441(1): 114167, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39004202

ABSTRACT

This research aims to explore the mechanism by which microRNAs may regulate the biological behavior of tumor cells in ALDH1+ fibrosarcoma. We identified differentially expressed miRNAs in ALDH + NMFH-1 cells, screened genes related to sarcoma metastasis in the TCGA database, and finally obtained key genes regulated by miRNAs that are involved in metastasis. The function and mechanism of these key genes were then validated at the cellular level. Using the ULCAN database, a significant correlation was found between hsa-mir-206 and mortality in sarcoma patients. WGCNA analysis identified 352 genes related to tumor metastasis. Through Venn diagrams, we obtained 15 metastasis-related genes regulated by hsa-mir-206. Survival analysis showed that SYNPO2 expression is significantly correlated with survival rate and is significantly underexpressed in multiple tumors. SYNPO2 showed a negative correlation with macrophages and a positive correlation with CD8+ T cells. After inhibiting the expression of hsa-mir-206 with siRNA plasmids, the mRNA expression of SYNPO2 was significantly upregulated. The results of CCK8 assay, scratch assay, and transwell assay showed that the proliferation and migration ability of NFMH-1 cells were promoted after SYNPO2 was inhibited. ALDH1+ tumor stem cells promote the proliferation and invasion of malignant fibrous histiocytoma cells by inhibiting SYNPO2 through hsa-mir-206.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , MicroRNAs , Neoplastic Stem Cells , Retinal Dehydrogenase , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Cell Proliferation/genetics , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Cell Movement/genetics , Cell Line, Tumor , Fibrosarcoma/pathology , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Disease Progression , Mice , Animals
4.
Br J Cancer ; 131(2): 325-333, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849477

ABSTRACT

BACKGROUND: We examined associations of CD44, CD24 and ALDH1A1 breast stem cell markers with mammographic breast density (MBD), a well-established breast cancer (BCa) risk factor. METHODS: We included 218 cancer-free women with biopsy-confirmed benign breast disease within the Nurses' Health Study (NHS) and NHSII. The data on BCa risk factors were obtained from biennial questionnaires. Immunohistochemistry (IHC) was done on tissue microarrays. For each core, the IHC expression was assessed using a semi-automated platform and expressed as percent of positively stained cells for each marker out of the total cell count. MBD was assessed with computer-assisted techniques. Generalised linear regression was used to examine the associations of each marker with square root-transformed percent density (PD), absolute dense and non-dense areas (NDA), adjusted for BCa risk factors. RESULTS: Stromal CD44 and ALDH1A1 expression was positively associated with PD (≥ 10% vs. <10% ß = 0.56, 95% confidence interval [CI] [0.06; 1.07] and ß = 0.81 [0.27; 1.34], respectively) and inversely associated with NDA (ß per 10% increase = -0.17 [-0.34; -0.01] and ß for ≥10% vs. <10% = -1.17 [-2.07; -0.28], respectively). Epithelial CD24 expression was inversely associated with PD (ß per 10% increase = -0.14 [-0.28; -0.01]. Stromal and epithelial CD24 expression was positively associated with NDA (ß per 10% increase = 0.35 [0.2 × 10-2; 0.70] and ß per 10% increase = 0.34 [0.11; 0.57], respectively). CONCLUSION: Expression of stem cell markers is associated with MBD.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Breast Density , CD24 Antigen , Hyaluronan Receptors , Retinal Dehydrogenase , Humans , Female , CD24 Antigen/metabolism , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/analysis , Aldehyde Dehydrogenase 1 Family/metabolism , Retinal Dehydrogenase/metabolism , Middle Aged , Adult , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/diagnostic imaging , Biopsy , Breast/pathology , Breast/diagnostic imaging , Breast/metabolism , Mammography/methods , Stem Cells/metabolism , Stem Cells/pathology , Biomarkers, Tumor/metabolism , Aldehyde Dehydrogenase/metabolism
5.
Funct Integr Genomics ; 24(3): 103, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38913281

ABSTRACT

Breast cancer severely affects women health. 70% of breast cancer are estrogen receptor positive. Breast cancer stem cells are a group of tumor with plasticity, causing tumor relapse and metastasis. RUNX3 is a tumor suppressor frequently inactivated in estrogen receptor positive breast cancer. However, the mechanism of how RUNX3 is involved in the regualation of cancer stem cell traits in estrogen receptor positive breast cancer remains elusive. In this study, we utilized cut-tag assay to investigate the binding profile RUNX3 in BT474 and T47D cell, and confirmed EXOSC4 as the bona-fide target of RUNX3; RUNX3 could bind to the promoter are of EXOSC4 to suppress its expression. Furthermore, EXOSC4 could increase the colony formation, cell invasion and mammosphere formation ability of breast cancer cells and upregulate the the expression of SOX2 and ALDH1. Consistent with these findings, EXOSC4 was associated with poorer survival for Luminal B/Her2 breast cancer patiens. At last, we confirmed that EXOSC4 mediated the tumor suppressive role of RUNX3 in breast cancer cells. In conclusion, we demonstrate that RUNX3 directly binds to the promoter region of EXOSC4, leading to the suppression of EXOSC4 expression and exerting a tumor-suppressive effect in estrogen receptor postivive breast cancer cells.


Subject(s)
Breast Neoplasms , Core Binding Factor Alpha 3 Subunit , Promoter Regions, Genetic , Female , Humans , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Retinal Dehydrogenase/metabolism , Retinal Dehydrogenase/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics
6.
Ecotoxicol Environ Saf ; 270: 115876, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38154155

ABSTRACT

Early life exposure to endocrine disrupting chemicals (EDCs) has been suggested to adversely affect reproductive health in humans and wildlife. Here, we characterize endocrine and adverse effects on the reproductive system after juvenile exposure to propiconazole (PROP) or imazalil (IMZ), two common azole fungicides with complex endocrine modes of action. Using the frog Xenopus tropicalis, two short-term (2-weeks) studies were conducted. I: Juveniles (2 weeks post metamorphosis (PM)) were exposed to 0, 17 or 178 µg PROP/L. II: Juveniles (6 weeks PM) were exposed to 0, 1, 12 or 154 µg IMZ/L. Histological analysis of the gonads revealed an increase in the number of dark spermatogonial stem cells (SSCs)/testis area, and in the ratio secondary spermatogonia: dark SSCs were increased in all IMZ groups compared to control. Key genes in gametogenesis, retinoic acid and sex steroid pathways were also analysed in the gonads. Testicular levels of 3ß-hsd, ddx4 were increased and cyp19 and id4 levels were decreased in the IMZ groups. In PROP exposed males, increased testicular aldh1a2 levels were detected, but no histological effects observed. Although no effects on ovarian histology were detected, ovarian levels of esr1, rsbn1 were increased in PROP groups, and esr1 levels were decreased in IMZ groups. In conclusion, juvenile azole exposure disrupted testicular expression of key genes in retinoic acid (PROP) and sex steroid pathways and in gametogenesis (IMZ). Our results further show that exposure to environmental concentrations of IMZ disrupted spermatogenesis in the juvenile testis, which is a cause for concern as it may lead to impaired fertility. Testicular levels of id4, ddx4 and the id4:ddx4 ratio were associated with the number of dark SSCs and secondary spermatogonia suggesting that they may serve as a molecular markers for disrupted spermatogenesis.


Subject(s)
Fungicides, Industrial , Humans , Male , Female , Animals , Fungicides, Industrial/metabolism , Xenopus laevis , Azoles/toxicity , Xenopus/metabolism , Testis , Spermatogenesis , Gonadal Steroid Hormones/metabolism , Tretinoin , Steroids/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/pharmacology , Retinal Dehydrogenase/metabolism
7.
J Obstet Gynaecol ; 44(1): 2361435, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39007780

ABSTRACT

BACKGROUND: Prognostic factors-based nomograms have been utilised to detect the likelihood of the specific cancer events. We have focused on the roles of aldehyde dehydrogenase 1 (ALDH1) and p-AKT in predicting the prognosis of BC patients. This study was designed to establish nomograms based on the integration of aldehyde dehydrogenase 1 (ALDH1) and p-AKT in predicting the disease-free survival (DFS) and overall survival (OS) of breast cancer (BC) patients. METHODS: Demographic and clinical data were obtained from BC patients admitted to our hospital between September 2015 and August 2016. Univariate and multivariate Cox regression analyses were utilised to analyse the risk factors of recurrence and mortality. The nomograms for predicting the DFS and OS were established using the screened risk factors. Stratified analysis was performed with the cut-off value of exp (pi) of 4.0-fold in DFS and OS, respectively. RESULTS: Multivariate Cox regression analysis indicated that ALDH, p-AKT and pathological stage III were independent risk factors for the recurrence among BC patients. ALDH1, p-AKT, pathological stage III and ER-/PR-/HER2- were independent risk factors for the mortality among BC patients. The established nomograms based on these factors were effective for predicting the DFS and OS with good agreement to the calibration curve and acceptable area under the receiver operating characteristic (ROC) curve. Finally, stratified analyses showed patients with a low pi showed significant decrease in the DFS and OS compared with those of high risk. CONCLUSION: We established nomograms for predicting the DFS and OS of BC patients based on ALDH1, p-AKT and pathological stages. The ER-/PR-/HER2- may be utilised to predict the OS rather than DFS in the BC patients.


Many breast cancer patients show poor response after treatment due to recurrence and metastasis. Therefore, early prediction of the disease-free survival and overall survival is crucial to the treatment outcome and clinical decision-making. In this study, we established nomograms with the demographic and clinical data from breast cancer patients admitted to our hospital between September 2015 and August 2016. Univariate and multivariate Cox regression analyses showed that some important proteins and signalling pathways were risk factors for decreased disease-free survival and overall survival of breast cancer patients. On this basis, we established an effective nomogram for predicting the disease-free survival and overall survival of these patients based on these factors. This study offers new options in the predicting the treatment outcome of breast cancer patients.


Subject(s)
Breast Neoplasms , Nomograms , Humans , Female , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Middle Aged , Disease-Free Survival , Adult , Risk Factors , Aldehyde Dehydrogenase 1 Family/metabolism , Neoplasm Recurrence, Local , Aged , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Retrospective Studies , Proportional Hazards Models , Biomarkers, Tumor/metabolism
8.
Biochem Biophys Res Commun ; 669: 85-94, 2023 08 20.
Article in English | MEDLINE | ID: mdl-37267864

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the cancer with the poorest prognosis. One of the major properties reflecting its poor prognosis is high-grade heterogeneity, which leads to insensitivity to anticancer treatments. Cancer stem cells (CSCs) acquire phenotypic heterogeneity, generating abnormally differentiated cells by asymmetric cell division. However, the detailed mechanism leading to phenotypic heterogeneity is largely unknown. Here, we showed that PDAC patients with co-upregulation of PKCλ and ALDH1A3 had the poorest clinical outcome. PKCλ knockdown by DsiRNA in the ALDH1high population of PDAC MIA-PaCa-2 cells attenuated the asymmetric distribution of the ALDH1A3 protein. To monitor asymmetric cell division of ALDH1A3-positive PDAC CSCs, we established stable Panc-1 PDAC clones expressing ALDH1A3-turboGFP (Panc-1-ALDH1A3-turboGFP cells). In addition to MIA-PaCa-2-ALDH1high cells, turboGFPhigh cells sorted from Panc-1-ALDH1A3-turboGFP cells showed asymmetric cell propagation of ALDH1A3 protein. PKCλ DsiRNA in Panc-1-ALDH1A3-turboGFP cells also attenuated the asymmetric distribution of ALDH1A3 protein. These results suggest that PKCλ regulates the asymmetric cell division of ALDH1A3-positive PDAC CSCs. Furthermore, Panc-1-ALDH1A3-turboGFP cells can be useful for the visualization and monitoring of CSC properties such as asymmetric cell division of ALDH1A3-positive PDAC CSCs in time-lapse imaging.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Asymmetric Cell Division , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Aldehyde Dehydrogenase 1 Family/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms
9.
FASEB J ; 36(4): e22224, 2022 04.
Article in English | MEDLINE | ID: mdl-35218575

ABSTRACT

Yes-associated protein (YAP), a central effector in the Hippo pathway, is involved in the regulation of organ size, stem cell self-renewal, and tissue regeneration. In this study, we observed YAP activation in patients with alcoholic steatosis, hepatitis, and cirrhosis. Accumulation of this protein in the nucleus was also observed in murine livers that were damaged after chronic-plus-single binge or moderate ethanol ingestion combined with carbon tetrachloride intoxication (ethanol/CCl4 ). To understand the role of this transcriptional coactivator in alcohol-related liver injury, we knocked out the Yap1 gene in hepatocytes of floxed homozygotes through adeno-associated virus (AAV8)-mediated deletion utilizing Cre recombinase. Yap1 hepatocyte-specific knockouts (KO) exhibited hemorrhage, massive hepatic necrosis, enhanced oxidative stress, elevated hypoxia, and extensive infiltration of CD11b+ inflammatory cells into hepatic microenvironments rich for connective tissue growth factor (Ctgf) during ethanol/CCl4 -induced liver damage. Analysis of whole-genome transcriptomics indicated upregulation of genes involved in hypoxia and extracellular matrix (ECM) remodeling, whereas genes related to hepatocyte proliferation, progenitor cell activation, and ethanol detoxification were downregulated in the damaged livers of Yap1 KO. Acetaldehyde dehydrogenase (Aldh)1a1, a gene that encodes a detoxification enzyme for aldehyde substrates, was identified as a potential YAP target because this gene could be transcriptionally activated by a hyperactive YAP mutant. The ectopic expression of the human ALDH1A1 gene caused increase in hepatocyte proliferation and decrease in hepatic necrosis, oxidative stress, ECM remodeling, and inflammation during ethanol/CCl4 -induced liver damage. Taken together, these observations indicated that YAP was crucial for liver repair during alcohol-associated injury. Its regulation of ALDH1A1 represents a new link in liver regeneration and detoxification.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Ethanol/toxicity , Liver Regeneration , Retinal Dehydrogenase/metabolism , YAP-Signaling Proteins/physiology , Aldehyde Dehydrogenase 1 Family/genetics , Animals , Cell Proliferation , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Dehydrogenase/genetics , Signal Transduction
10.
Am J Med Genet A ; 191(1): 90-99, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36263470

ABSTRACT

Aldehyde Dehydrogenase 1, Family Member A2 (ALDH1A2) is essential for the synthesis of retinoic acid from vitamin A. Studies in model organisms demonstrate a critical role for ALDH1A2 in embryonic development, yet few pathogenic variants are linked to congenital anomalies in humans. We present three siblings with multiple congenital anomaly syndrome linked to biallelic sequence variants in ALDH1A2. The major congenital malformations affecting these children include tetralogy of Fallot, absent thymus, diaphragmatic eventration, and talipes equinovarus. Upper airway anomalies, hypocalcemia, and dysmorphic features are newly reported in this manuscript. In vitro functional validation of variants indicated that substitutions reduced the expression of the enzyme. Our clinical and functional data adds to a recent report of biallelic ALDH1A2 pathogenic variants in two families with a similar constellation of congenital malformations. These findings provide further evidence for an autosomal recessive ALDH1A2-deficient recognizable malformation syndrome involving the diaphragm, cardiac and musculoskeletal systems.


Subject(s)
Tretinoin , Child , Humans , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Tretinoin/metabolism , Retinal Dehydrogenase/genetics
11.
J Pathol ; 256(3): 349-362, 2022 03.
Article in English | MEDLINE | ID: mdl-34897678

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) is an aggressive pediatric cancer with poor prognosis. Cancer stem cells (CSCs) are seeds for tumor relapse and metastasis. However, pathways that maintain stemness genes are not fully understood. Here, we report that the enzyme euchromatic histone lysine methyltransferase 1 (EHMT1) is expressed in primary and relapse ARMS tumors. EHMT1 suppression impaired motility and induced differentiation in ARMS cell lines and reduced tumor progression in a mouse xenograft model in vivo. RNA sequencing of EHMT1-depleted cells revealed downregulation of ALDH1A1 that is associated with CSCs. Consistent with this, inhibition of ALDH1A1 expression and activity mimicked EHMT1 depletion phenotypes and reduced tumorsphere formation. Mechanistically, we demonstrate that EHMT1 does not bind to the ALDH1A1 promoter but activates it by stabilizing C/EBPß, a known regulator of ALDH1A1 expression. Our findings identify a role for EHMT1 in maintenance of stemness by regulating ALDH1A1 expression and suggest that targeting ALDH+ cells is a promising strategy in ARMS. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neoplastic Stem Cells/enzymology , Retinal Dehydrogenase/metabolism , Rhabdomyosarcoma, Alveolar/enzymology , Aldehyde Dehydrogenase 1 Family/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Phenotype , Retinal Dehydrogenase/genetics , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Signal Transduction , Tumor Burden
12.
Int J Mol Sci ; 24(3)2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36768723

ABSTRACT

Recurrent disease and treatment-associated chemoresistance are the two main factors accounting for poor clinical outcomes of ovarian cancer (OC) patients. Both can be associated with cancer stem cells (CSCs), which contribute to cancer formation, progression, chemoresistance, and recurrence. Hence, this study investigated whether the expression of known CSC-associated markers ALDH1A, CD44, and CD133 may predict OC patient prognosis. We analyzed their expression in primary epithelial ovarian cancer (EOC) patients using immunohistochemistry and related them to clinicopathological data, including overall survival (OS) and progression-free survival (PFS). Expression of ALDH1A1 was detected in 32%, CD133 in 28%, and CD44 in 33% of cases. While Kaplan-Meier analysis revealed no association of the expression of CD133 and CD44 with PFS and OS, ALDH1A1-positive patients were characterized with both significantly shorter OS (p = 0.00022) and PFS (p = 0.027). Multivariate analysis demonstrated that the expression of ALDH1A1, FIGO stage III-IV, and residual disease after suboptimal debulking or neoadjuvant chemotherapy correlated with shorter OS. The results of this study identify ALDH1A1 as a potential independent prognostic factor of shorter OS and PFS in EOC patients. Therefore, targeting ALDH1A1-positive cancer cells may be a promising therapeutic strategy to influence the disease course and treatment response.


Subject(s)
Hyaluronan Receptors , Ovarian Neoplasms , Female , Humans , Aldehyde Dehydrogenase 1 Family/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial/pathology , Follow-Up Studies , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Prognosis , Retinal Dehydrogenase/metabolism
13.
Biochem Biophys Res Commun ; 628: 141-146, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36084552

ABSTRACT

Aldehyde dehydrogenase 1A1 (ALDH1A1) is an enzyme that catalyzes the NAD+-dependent oxidation of aldehydes to carboxylic acids, participating in various metabolic processes. Currently, only structures from human and Ovis aries have been reported. Here we show a 2.89 Å resolution structure of ALDH1A1 from mice using X-ray crystallography. We performed a detailed analysis of the structure and compared it with ALDH1A1 structures from two other species, highlighting the significance of the differences. Structural superimposition reveals that the tetrameric molecule is asymmetrical, and the NAD+-binding domain exhibits a certain rotation. In addition, the noticeable structural differences were detected, including the unique contact between Ser461 and Asp148, as well as the side chain orientations of three amino acids residues, Asn474, Met471 and Phe466. This study helps to expand the structural diversity of the ALDH family.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase , NAD , Retinal Dehydrogenase , Aldehyde Dehydrogenase/chemistry , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family/chemistry , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehydes/metabolism , Amino Acids , Animals , Carboxylic Acids , Crystallography, X-Ray , Mice , NAD/metabolism , Retinal Dehydrogenase/chemistry , Retinal Dehydrogenase/metabolism
14.
Proc Natl Acad Sci U S A ; 116(41): 20679-20688, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548432

ABSTRACT

The presence of progenitor or stem cells in the adult pancreas and their potential involvement in homeostasis and cancer development remain unresolved issues. Here, we show that mouse centroacinar cells can be identified and isolated by virtue of the mitochondrial enzyme Aldh1b1 that they uniquely express. These cells are necessary and sufficient for the formation of self-renewing adult pancreatic organoids in an Aldh1b1-dependent manner. Aldh1b1-expressing centroacinar cells are largely quiescent, self-renew, and, as shown by genetic lineage tracing, contribute to all 3 pancreatic lineages in the adult organ under homeostatic conditions. Single-cell RNA sequencing analysis of these cells identified a progenitor cell population, established its molecular signature, and determined distinct differentiation pathways to early progenitors. A distinct feature of these progenitor cells is the preferential expression of small GTPases, including Kras, suggesting that they might be susceptible to Kras-driven oncogenic transformation. This finding and the overexpression of Aldh1b1 in human and mouse pancreatic cancers, driven by activated Kras, prompted us to examine the involvement of Aldh1b1 in oncogenesis. We demonstrated genetically that ablation of Aldh1b1 completely abrogates tumor development in a mouse model of KrasG12D-induced pancreatic cancer.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/pathology , Mutation , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Stem Cells/pathology , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction , Single-Cell Analysis , Stem Cells/metabolism
15.
J Obstet Gynaecol Res ; 48(4): 1011-1018, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35194895

ABSTRACT

AIM: Integrin alpha 7 (ITGA7) regulates cancer stemness and metastasis in several malignancies, while its role in cervical cancer is obscure. Therefore, the current study aimed to investigate the correlation among ITGA7, cluster of differentiation 133 (CD133), and aldehyde dehydrogenase isoform 1 (ALDH1), as well as their relation to tumor features and survival in cervical cancer patients. METHODS: A total of 133 surgical cervical cancer patients were enrolled. Tumor ITGA7, CD133, and ALDH1 expressions were determined by immunohistochemistry (IHC). Furthermore, the clinicopathological features, disease-free survival (DFS), and overall survival (OS) were collected. RESULTS: ITGA7 expression positively related to CD133 expression (p = 0.040) and ALDH1 expression (p < 0.001). Besides, ITGA7 (p = 0.001), CD133 (p = 0.016), and ALDH1 (p = 0.009) high expressions linked with poor tumor differentiation; meanwhile, ITGA7 (p = 0.010) and ALDH1 (p = 0.004) high expressions correlated with more prevalence of lymph node metastasis. However, ITGA7, CD133, or ALDH1 expression was not associated with other clinicopathological features. Inspiringly, it was worth noting that ITGA7 (p = 0.009), CD133 (p = 0.041), and ALDH1 (p = 0.035) high expressions predicted unfavorable DFS; meanwhile, both ITGA7 (p = 0.021) and ALDH1 (p = 0.023) high expressions but not CD133 expression (p = 0.169) forecasted exasperated OS. CONCLUSION: ITGA7, CD133, ALDH1 are inter-correlated, and linked with poor differentiation, lymph node metastasis as well as worse survival in surgical cervical cancer.


Subject(s)
AC133 Antigen , Aldehyde Dehydrogenase 1 Family , Integrins , Uterine Cervical Neoplasms , AC133 Antigen/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Female , Humans , Integrins/metabolism , Lymphatic Metastasis/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Uterine Cervical Neoplasms/pathology
16.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162954

ABSTRACT

Mesothelin (MSLN) overexpression (OE) is a frequent finding in ovarian carcinomas and increases cell survival and tumor aggressiveness. Since cancer stem cells (CSCs) contribute to pathogenesis, chemoresistance and malignant behavior in ovarian cancer (OC), we hypothesized that MSLN expression could be creating a favorable environment that nurtures CSCs. In this study, we analyzed the expression of MSLN and CSC markers SOX2 and ALDH1 by immunohistochemistry (IHC) in different model systems: primary high-grade serous carcinomas (HGSCs) and OC cell lines, including cell lines that were genetically engineered for MSLN expression by either CRISPR-Cas9-mediated knockout (Δ) or lentivirus-mediated OE. Cell lines, wild type and genetically engineered, were evaluated in 2D and 3D culture conditions and xenografted in nude mice. We observed that MSLN was widely expressed in HGSC, and restricted expression was observed in OC cell lines. In contrast, SOX2 and ALDH1 expression was limited in all tissue and cell models. Most importantly, the expression of CSC markers was independent of MSLN expression, and manipulation of MSLN expression did not affect CSC markers. In conclusion, MSLN expression is not involved in driving the CSC phenotype.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Cystadenocarcinoma, Serous/pathology , Mesothelin/metabolism , Ovarian Neoplasms/pathology , Retinal Dehydrogenase/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cystadenocarcinoma, Serous/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Retrospective Studies , Xenograft Model Antitumor Assays
17.
Molecules ; 27(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36500483

ABSTRACT

Folate (vitamin B9) is involved in one-carbon transfer reactions and plays a significant role in nucleic acid synthesis and control of cellular proliferation, among other key cellular processes. It is now recognized that the role of folates in different stages of carcinogenesis is complex, and more research is needed to understand how folate reactions become dysregulated in cancers and the metabolic consequences that occur as a result. ALDH1L1 (cytosolic 10-formyltetrahydrofolate dehydrogenase), an enzyme of folate metabolism expressed in many tissues, is ubiquitously downregulated in cancers and is not expressed in cancer cell lines. The RT4 cell line (derived from papillary bladder cancer) which expresses high levels of ALDH1L1 represents an exception, providing an opportunity to explore the metabolic consequences of the loss of this enzyme. We have downregulated this protein in RT4 cells (shRNA driven knockdown or CRISPR driven knockout) and compared metabolomes of ALDH1L1-expressing and -deficient cells to determine if metabolic changes linked to the loss of this enzyme might provide proliferative and/or survival advantages for cancer cells. In this study, cell extracts were analyzed using Ultra High Performance Liquid Chromatography High Resolution Mass Spectrometry (UHPLC-HR-MS). A total of 13,339 signals were identified or annotated using an in-house library and public databases. Supervised and unsupervised multivariate analysis revealed metabolic differences between RT4 cells and ALDH1L1-deficient clones. Glycine (8-fold decrease) and metabolites derived from S-adenosylmethionine utilizing pathways were significantly decreased in the ALDH1L1-deficient clones, compared with RT4 cells. Other changes linked to ALDH1L1 downregulation include decreased levels of amino acids, Krebs cycle intermediates, and ribose-5-phosphate, and increased nicotinic acid. While the ALDH1L1-catalyzed reaction is directly linked to glycine biosynthesis and methyl group flux, its overall effect on cellular metabolism extends beyond immediate metabolic pathways controlled by this enzyme.


Subject(s)
Folic Acid , Neoplasms , Humans , Folic Acid/metabolism , Glycine/metabolism , Retinal Dehydrogenase/metabolism , Methylation , Aldehyde Dehydrogenase 1 Family/metabolism , S-Adenosylmethionine/metabolism , Metabolomics
18.
Molecules ; 27(2)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35056791

ABSTRACT

Aldehyde dehydrogenase-1a1 (ALDH1a1), the enzyme responsible for the oxidation of retinal into retinoic acid, represents a key therapeutic target for the treatment of debilitating disorders such as cancer, obesity, and inflammation. Drugs that can inhibit ALDH1a1 include disulfiram, an FDA-approved drug to treat chronic alcoholism. Disulfiram, by carbamylation of the catalytic cysteines, irreversibly inhibits ALDH1a1 and ALDH2. The latter is the isozyme responsible for important physiological processes such as the second stage of alcohol metabolism. Given the fact that ALDH1a1 has a larger substrate tunnel than that in ALDH2, replacing disulfiram ethyl groups with larger motifs will yield selective ALDH1a1 inhibitors. We report herein the synthesis of new inhibitors of ALDH1a1 where (hetero)aromatic rings were introduced into the structure of disulfiram. Most of the developed compounds retained the anti-ALDH1a1 activity of disulfiram; however, they were completely devoid of inhibitory activity against ALDH2.


Subject(s)
Acetaldehyde Dehydrogenase Inhibitors/chemistry , Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Disulfiram/chemistry , Disulfiram/pharmacology , Retinal Dehydrogenase/antagonists & inhibitors , Acetaldehyde Dehydrogenase Inhibitors/chemical synthesis , Acetaldehyde Dehydrogenase Inhibitors/metabolism , Aldehyde Dehydrogenase 1 Family/chemistry , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial/chemistry , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Disulfiram/analogs & derivatives , Disulfiram/chemical synthesis , Humans , Molecular Docking Simulation , Recombinant Proteins/metabolism , Retinal Dehydrogenase/chemistry , Retinal Dehydrogenase/metabolism
19.
Hum Genet ; 140(10): 1471-1485, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34417872

ABSTRACT

Argininosuccinate lyase (ASL) is essential for the NO-dependent regulation of tyrosine hydroxylase (TH) and thus for catecholamine production. Using a conditional mouse model with loss of ASL in catecholamine neurons, we demonstrate that ASL is expressed in dopaminergic neurons in the substantia nigra pars compacta, including the ALDH1A1 + subpopulation that is pivotal for the pathogenesis of Parkinson disease (PD). Neuronal loss of ASL results in catecholamine deficiency, in accumulation and formation of tyrosine aggregates, in elevation of α-synuclein, and phenotypically in motor and cognitive deficits. NO supplementation rescues the formation of aggregates as well as the motor deficiencies. Our data point to a potential metabolic link between accumulations of tyrosine and seeding of pathological aggregates in neurons as initiators for the pathological processes involved in neurodegeneration. Hence, interventions in tyrosine metabolism via regulation of NO levels may be therapeutic beneficial for the treatment of catecholamine-related neurodegenerative disorders.


Subject(s)
Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/metabolism , Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Animals , Disease Models, Animal , Humans , Mice , Phenotype , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism
20.
Bioconjug Chem ; 32(2): 234-238, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33502173

ABSTRACT

Aldehyde dehydrogenase 1 (ALDH1) plays an important role as a stem cell marker. In the field of stem cell biology, a green fluorescent ALDH1 probe has been principally used, but there is a need for more options in probe color. We designed and synthesized two blue fluorescent ALDH1 probes using 8-amino BODIPY and aminomethylbenzaldehyde. These probes can be simultaneously used with other color probes. Here, we demonstrate successful examples of the simultaneous use of these probes with green fluorescent protein.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Boron Compounds/chemistry , Fluorescent Dyes/chemistry , Amines/chemistry , Cell Line, Tumor , Humans
SELECTION OF CITATIONS
SEARCH DETAIL