1
|
Li S, Li H, Bennewitz K, Poschet G, Buettner M, Hausser I, Szendroedi J, Nawroth PP, Kroll J. Combined loss of glyoxalase 1 and aldehyde dehydrogenase 3a1 amplifies dicarbonyl stress, impairs proteasome activity resulting in hyperglycemia and activated retinal angiogenesis. Metabolism 2025:156149. [PMID: 39892865 DOI: 10.1016/j.metabol.2025.156149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND & AIMS Any energy consumption results in the generation of highly reactive dicarbonyls and the need to prevent excessive dicarbonyls accumulation through the activity of several interdependent detoxification enzymes. Glyoxalase 1 (GLO1) knockout zebrafish showed only moderately elevated methylglyoxal (MG) levels, but increased Aldehyde Dehydrogenases (ALDH) activity and increased aldh3a1 expression. Elevated levels of 4-hydroxynonenal (4-HNE) but no MG increase were observed in ALDH3A1KO. The question of whether ALDH3A1 prevents MG formation as a compensatory mechanism in the absence of GLO1 remained unclear. METHODS To investigate whether ALDH3A1 detoxifies MG as a compensatory mechanism in the absence of GLO1, the GLO1/ALDH3A1 double knockout (DKO) zebrafish was first generated. Various metabolites including advanced glycation end products (AGEs), as well as glucose metabolism and hyaloid vasculature were analyzed in GLO1KO, ALDH3A1KO and GLO1/ALDH3A1DKO zebrafish. RESULTS In the absence of GLO1 and ALDH3A1, MG-H1 levels were increased. MG-H1 accumulation led to a severe deterioration of proteasome function, resulting in impaired glucose homeostasis and consequently amplified angiogenic activation of the hyaloid and retinal vasculature. Rescue of these pathological processes could be observed by using L-carnosine, and proteasome activator betulinic acid. CONCLUSION The present data, together with previous studies, suggest that ALDH3A1 and GLO1 are important detoxification enzymes that prevent the deleterious effects of MG-H1 accumulation on proteasome function, glucose homeostasis and vascular function.
Collapse
Affiliation(s)
- Shu Li
- Department of Vascular Biology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Hao Li
- German Cancer Research Center (DKFZ), Unit D400, Heidelberg 69120, Germany
| | - Katrin Bennewitz
- Department of Vascular Biology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany
| | - Michael Buettner
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany
| | - Ingrid Hausser
- Institute of Pathology IPH, EM Lab, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Julia Szendroedi
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Peter Paul Nawroth
- Medical Clinic and Polyclinic II, University Hospital Dresden, Dresden 01307, Germany
| | - Jens Kroll
- Department of Vascular Biology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany.
| |
Collapse
|
2
|
Kong S, Pan H, Zhang YW, Wang F, Chen J, Dong J, Yin C, Wu J, Zhou D, Peng J, Ma J, Zhou J, Ge D, Lu Y, Wei DD, Fang J, Han W, Shen C, Koeffler HP, Wang B, Jiang Y, Jiang YY. Targeting aldehyde dehydrogenase ALDH3A1 increases ferroptosis vulnerability in squamous cancer. Oncogene 2025:10.1038/s41388-025-03277-4. [PMID: 39863749 DOI: 10.1038/s41388-025-03277-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/15/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Ferroptosis is a unique modality of regulated cell death induced by excessive lipid peroxidation, playing a crucial role in tumor suppression and providing potential therapeutic strategy for cancer treatment. Here, we find that aldehyde dehydrogenase-ALDH3A1 tightly links to ferroptosis in squamous cell carcinomas (SCCs). Functional assays demonstrate the enzymatic activity-dependent regulation of ALDH3A1 in protecting SCC cells against ferroptosis through catalyzing aldehydes and mitigating lipid peroxidation. Furthermore, a specific covalent inhibitor of ALDH3A1-EN40 significantly enhances the ferroptosis sensitivity induced by the ferroptosis inducer. The combination of EN40 and a ferroptosis inducer exhibits a synergistic effect, effectively inhibiting the proliferation of SCC cells/organoids and suppressing tumor growth both in vitro and in vivo. On mechanism, high expression of ALDH3A1 is transcriptionally governed by TP63, which binds to super-enhancer of ALDH3A1. Collectively, our findings reveal a yet-unrecognized function of ALDH3A1 exploited by SCC cells to evade ferroptosis, and targeting ALDH3A1 may enhance the effect of ferroptosis-induced therapy in SCCs.
Collapse
Affiliation(s)
- Shuai Kong
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Huaguang Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yuan-Wei Zhang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Fei Wang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Jian Chen
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jinxiu Dong
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Chuntong Yin
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Jiaqi Wu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Dan Zhou
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jingyi Peng
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Junboya Ma
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Jianian Zhou
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Dianlong Ge
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yan Lu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Dan-Dan Wei
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jinman Fang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wei Han
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Chengyin Shen
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - H Phillip Koeffler
- Department of Medicine, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Boshi Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| | - Yuan Jiang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- University of Science and Technology of China, Hefei, 230026, China.
| | - Yan-Yi Jiang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
3
|
Li A, Dong L, Li X, Yi J, Ma J, Zhou J. ALDH3A1-mediated detoxification of reactive aldehydes contributes to distinct muscle responses to denervation and Amyotrophic Lateral Sclerosis progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626422. [PMID: 39677625 PMCID: PMC11642873 DOI: 10.1101/2024.12.02.626422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Different muscles exhibit varied susceptibility to degeneration in Amyotrophic Lateral Sclerosis (ALS), a fatal neuromuscular disorder. Extraocular muscles (EOMs) are particularly resistant to ALS progression and exploring the underlying molecular nature may deliver great therapeutic value. Reactive aldehyde 4-hydroxynonenal (HNE) is implicated in ALS pathogenesis and ALDH3A1 is an inactivation-resistant intracellular detoxifier of 4-HNE protecting eyes against UV-induced oxidative stress. Here we detected prominently higher levels of ALDH3A1 in mouse EOMs than other muscles under normal physiological conditions. In an ALS mouse model (hSOD1G93A) reaching end-stage, ALDH3A1 expression was sustained at high level in EOMs, whereas substantial upregulation of ALDH3A1 occurred in soleus and diaphragm. The upregulation was less pronounced in extensor digitorum longus (EDL) muscle, which endured the most severe pathological remodeling as demonstrated by unparalleled upregulation of a denervation marker ANKRD1 expression. Interestingly, sciatic nerve transection in wildtype mice induced ALDH3A1 and ANKRD1 expression in an inverse manner over muscle type and time. Adeno-associated virus enforced overexpression of ALDH3A1 protected myotubes from 4-HNE-induced DNA fragmentation, plasma membrane leakage and restored MG53-mediated membrane repair. Our data indicate that ALDH3A1 may contribute to distinct muscle resistance to ALS through detoxifying reactive aldehydes.
Collapse
Affiliation(s)
- Ang Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, TX, 76019, USA
| | - Li Dong
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, TX, 76019, USA
| | - Xuejun Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, TX, 76019, USA
| | - Jianxun Yi
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, TX, 76019, USA
| | - Jianjie Ma
- Department of Surgery, Division of Surgical Sciences, University of Virginia, Charlottesville, VA, 22903, USA
| | - Jingsong Zhou
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, TX, 76019, USA
| |
Collapse
|
4
|
Zhang Y, Chen XY, Hu YZ, Zhang X, Zheng SF, Hu SS. Application of transgenic mice to the molecular pathogenesis of cataract. Int J Ophthalmol 2024; 17:1929-1948. [PMID: 39430018 PMCID: PMC11422363 DOI: 10.18240/ijo.2024.10.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/29/2024] [Indexed: 10/22/2024] Open
Abstract
One of the most prevalent disorders that cause blindness worldwide is cataract, and its essence is the visual disorder caused by the opacity of the lens. The significant degree of variation in cataracts and the fact that a variety of factors can impact a patient's lens transparency make it especially crucial to investigate the pathogenesis of cataracts at the molecular level. It has been found that more than 60 genes are linked to the formation of cataracts, and the construction of a transgenic mouse model of cataract similar to the selection of human lens clouding due to a variety of causes has become an important means of studying the pathogenesis of cataract. Therefore, the research on the application of transgenic mice to the molecular pathogenesis of cataracts will be the main topic of this review of the literature.
Collapse
Affiliation(s)
- Yue Zhang
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| | - Xiao-Ya Chen
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| | - Yu-Zhu Hu
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| | - Xiao Zhang
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| | - Shun-Fei Zheng
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| | - Shan-Shan Hu
- Department of Ophthalmology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, China
| |
Collapse
|
5
|
Amodei L, Ruggieri AG, Potenza F, Viele M, Dufrusine B, Franciotti R, Pietrangelo L, Ardini M, Stuppia L, Federici L, De Laurenzi V, Sallese M. Sil1-deficient fibroblasts generate an aberrant extracellular matrix leading to tendon disorganisation in Marinesco-Sjögren syndrome. J Transl Med 2024; 22:787. [PMID: 39180052 PMCID: PMC11342654 DOI: 10.1186/s12967-024-05582-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Marinesco-Sjögren syndrome (MSS) is an autosomal recessive neuromuscular disorder that arises in early childhood and is characterized by congenital cataracts, myopathy associated with muscle weakness, and degeneration of Purkinje neurons leading to ataxia. About 60% of MSS patients have loss-of-function mutations in the SIL1 gene. Sil1 is an endoplasmic reticulum (ER) protein required for the release of ADP from the master chaperone Bip, which in turn will release the folded proteins. The expression of non-functional Sil1 leads to the accumulation of unfolded proteins in the ER and this triggers the unfolded protein response (UPR). A dysfunctional UPR could be a key element in the pathogenesis of MSS, although our knowledge of the molecular pathology of MSS is still incomplete. METHODS RNA-Seq transcriptomics was analysed using the String database and the Ingenuity Pathway Analysis platform. Fluorescence confocal microscopy was used to study the remodelling of the extracellular matrix (ECM). Transmission electron microscopy (TEM) was used to reveal the morphology of the ECM in vitro and in mouse tendon. RESULTS Our transcriptomic analysis, performed on patient-derived fibroblasts, revealed 664 differentially expressed (DE) transcripts. Enrichment analysis of DE genes confirmed that the patient fibroblasts have a membrane trafficking issue. Furthermore, this analysis indicated that the extracellular space/ECM and the cell adhesion machinery, which together account for around 300 transcripts, could be affected in MSS. Functional assays showed that patient fibroblasts have a reduced capacity of ECM remodelling, reduced motility, and slower spreading during adhesion to Petri dishes. TEM micrographs of negative-stained ECM samples from these fibroblasts show differences of filaments in terms of morphology and size. Finally, structural analysis of the myotendinous junction of the soleus muscle and surrounding regions of the Achilles tendon revealed a disorganization of collagen fibres in the mouse model of MSS (woozy). CONCLUSIONS ECM alterations can affect the proper functioning of several organs, including those damaged in MSS such as the central nervous system, skeletal muscle, bone and lens. On this basis, we propose that aberrant ECM is a key pathological feature of MSS and may help explain most of its clinical manifestations.
Collapse
Affiliation(s)
- Laura Amodei
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Anna Giulia Ruggieri
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Francesca Potenza
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Marianna Viele
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Beatrice Dufrusine
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, 64100, Italy
| | | | | | - Matteo Ardini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Psychological Health and Territorial Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Luca Federici
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
| | - Michele Sallese
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy.
- Center for Advanced Studies and Technology (CAST), Chieti, Italy.
| |
Collapse
|
6
|
Duan X, Hu H, Wang L, Chen L. Aldehyde dehydrogenase 1 family: A potential molecule target for diseases. Cell Biol Int 2024. [PMID: 38800962 DOI: 10.1002/cbin.12188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024]
Abstract
Aldehyde dehydrogenase 1 (ALDH1), a crucial aldehyde metabolizing enzyme, has six family members. The ALDH1 family is expressed in various tissues, with a significant presence in the liver. It plays a momentous role in several pathophysiological processes, including aldehyde detoxification, oxidative stress, and lipid peroxidation. Acetaldehyde detoxification is the fundamental function of the ALDH1 family in participating in vital pathological mechanisms. The ALDH1 family can catalyze retinal to retinoic acid (RA) that is a hormone-signaling molecule and plays a vital role in the development and adult tissues. Furthermore, there is a need for further and broader research on the role of the ALDH1 family as a signaling molecule. The ALDH1 family is widely recognized as a cancer stem cell (CSC) marker and plays a significant role in the proliferation, invasion, metastasis, prognosis, and drug resistance of cancer. The ALDH1 family also participates in other human diseases, such as neurodegenerative diseases, osteoarthritis, diabetes, and atherosclerosis. It can inhibit disease progression by inhibiting/promoting the expression/activity of the ALDH1 family. In this review, we comprehensively analyze the tissue distribution, and functions of the ALDH1 family. Additionally, we review the involvement of the ALDH1 family in diseases, focusing on the underlying pathological mechanisms and briefly talk about the current status and development of ALDH1 family inhibitors. The ALDH1 family presents new possibilities for treating diseases, with both its upstream and downstream pathways serving as promising targets for therapeutic intervention. This offers fresh perspectives for drug development in the field of disease research.
Collapse
Affiliation(s)
- Xiangning Duan
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| | - Haoliang Hu
- Changde Research Centre for Artificial Intelligence and Biomedicine, Zoology Key Laboratory of Hunan Higher Education, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
| | - Lingzhi Wang
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou, Hunan, China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Duan JJ, Cai J, Gao L, Yu SC. ALDEFLUOR activity, ALDH isoforms, and their clinical significance in cancers. J Enzyme Inhib Med Chem 2023; 38:2166035. [PMID: 36651035 PMCID: PMC9858439 DOI: 10.1080/14756366.2023.2166035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
High aldehyde dehydrogenase (ALDH) activity is a metabolic feature of adult stem cells and various cancer stem cells (CSCs). The ALDEFLUOR system is currently the most commonly used method for evaluating ALDH enzyme activity in viable cells. This system is applied extensively in the isolation of normal stem cells and CSCs from heterogeneous cell populations. For many years, ALDH1A1 has been considered the most important subtype among the 19 ALDH family members in determining ALDEFLUOR activity. However, in recent years, studies of many types of normal and tumour tissues have demonstrated that other ALDH subtypes can also significantly influence ALDEFLUOR activity. In this article, we briefly review the relationships between various members of the ALDH family and ALDEFLUOR activity. The clinical significance of these ALDH isoforms in different cancers and possible directions for future studies are also summarised.
Collapse
Affiliation(s)
- Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital; Third Military Medical University (Army Medical University), Chongqing, China,International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China,Ministry of Education, Key Laboratory of Cancer Immunopathology, Chongqing, China
| | - Jiao Cai
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital; Third Military Medical University (Army Medical University), Chongqing, China,International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China,Ministry of Education, Key Laboratory of Cancer Immunopathology, Chongqing, China
| | - Lei Gao
- Department of Hematology, Xinqiao Hospital; Third Medical University (Army Medical University), Chongqing, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital; Third Military Medical University (Army Medical University), Chongqing, China,International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, China,Ministry of Education, Key Laboratory of Cancer Immunopathology, Chongqing, China,Jin-feng Laboratory, Chongqing, China,CONTACT Shi-Cang Yu Department of Stem Cell and Regenerative Medicine, Third Military Medical University (Army Medical University), Chongqing400038, China
| |
Collapse
|
8
|
Kundu B, Iyer MR. A patent review on aldehyde dehydrogenase inhibitors: an overview of small molecule inhibitors from the last decade. Expert Opin Ther Pat 2023; 33:651-668. [PMID: 38037334 DOI: 10.1080/13543776.2023.2287515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
INTRODUCTION Physiological and pathophysiological effects arising from detoxification of aldehydes in humans implicate the enzyme aldehyde dehydrogenase (ALDH) gene family comprising of 19 isoforms. The main function of this enzyme family is to metabolize reactive aldehydes to carboxylic acids. Dysregulation of ALDH activity has been associated with various diseases. Extensive research has since gone into studying ALHD isozymes, their structural biology and developing small-molecule inhibitors. Novel chemical strategies to enhance the selectivity of ALDH inhibitors have now appeared. AREAS COVERED A comprehensive review of patent literature related to aldehyde dehydrogenase inhibitors in the last decade and half (2007-2022) is provided. EXPERT OPINION Aldehyde dehydrogenase (ALDH) is an important enzyme that metabolizes reactive exogenous and endogenous aldehydes in the body through NAD(P)±dependent oxidation. Hence this family of enzymes possess important physiological as well as toxicological roles in human body. Significant efforts in the field have led to potent inhibitors with approved clinical agents for alcohol use disorder therapy. Further clinical translation of novel compounds targeting ALDH inhibition will validate the promised therapeutic potential in treating many human diseases.The scientific/patent literature has been searched on SciFinder-n, Reaxys, PubMed, Espacenet and Google Patents. The search terms used were 'ALDH inhibitors', 'Aldehyde Dehydrogenase Inhibitors'.
Collapse
Affiliation(s)
- Biswajit Kundu
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
9
|
Xanthis V, Mantso T, Dimtsi A, Pappa A, Fadouloglou VE. Human Aldehyde Dehydrogenases: A Superfamily of Similar Yet Different Proteins Highly Related to Cancer. Cancers (Basel) 2023; 15:4419. [PMID: 37686694 PMCID: PMC10650815 DOI: 10.3390/cancers15174419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The superfamily of human aldehyde dehydrogenases (hALDHs) consists of 19 isoenzymes which are critical for several physiological and biosynthetic processes and play a major role in the organism's detoxification via the NAD(P) dependent oxidation of numerous endogenous and exogenous aldehyde substrates to their corresponding carboxylic acids. Over the last decades, ALDHs have been the subject of several studies as it was revealed that their differential expression patterns in various cancer types are associated either with carcinogenesis or promotion of cell survival. Here, we attempt to provide a thorough review of hALDHs' diverse functions and 3D structures with particular emphasis on their role in cancer pathology and resistance to chemotherapy. We are especially interested in findings regarding the association of structural features and their changes with effects on enzymes' functionalities. Moreover, we provide an updated outline of the hALDHs inhibitors utilized in experimental or clinical settings for cancer therapy. Overall, this review aims to provide a better understanding of the impact of ALDHs in cancer pathology and therapy from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | - Vasiliki E. Fadouloglou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
10
|
Kwong JMK, Caprioli J, Lee JCY, Song Y, Yu FJ, Bian J, Sze YH, Li KK, Do CW, To CH, Lam TC. Differential Responses of Retinal Neurons and Glia Revealed via Proteomic Analysis on Primary and Secondary Retinal Ganglion Cell Degeneration. Int J Mol Sci 2023; 24:12109. [PMID: 37569482 PMCID: PMC10418669 DOI: 10.3390/ijms241512109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
To explore the temporal profile of retinal proteomes specific to primary and secondary retinal ganglion cell (RGC) loss. Unilateral partial optic nerve transection (pONT) was performed on the temporal side of the rat optic nerve. Temporal and nasal retinal samples were collected at 1, 4 and 8 weeks after pONT (n = 4 each) for non-biased profiling with a high-resolution hybrid quadrupole time-of-flight mass spectrometry running on label-free SWATHTM acquisition (SCIEX). An information-dependent acquisition ion library was generated using ProteinPilot 5.0 and OneOmics cloud bioinformatics. Combined proteome analysis detected 2531 proteins with a false discovery rate of <1%. Compared to the nasal retina, 10, 25 and 61 significantly regulated proteins were found in the temporal retina at 1, 4, and 8 weeks, respectively (p < 0.05, FC ≥ 1.4 or ≤0.7). Eight proteins (ALDH1A1, TRY10, GFAP, HBB-B1, ALB, CDC42, SNCG, NEFL) were differentially expressed for at least two time points. The expressions of ALDH1A1 and SNCG at nerve fibers were decreased along with axonal loss. Increased ALDH1A1 localization in the inner nuclear layer suggested stress response. Increased GFAP expression demonstrated regional reactivity of astrocytes and Muller cells. Meta-analysis of gene ontology showed a pronounced difference in endopeptidase and peptidase inhibitor activity. Temporal proteomic profiling demonstrates established and novel protein targets associated with RGC damage.
Collapse
Affiliation(s)
- Jacky M. K. Kwong
- Ophthalmology, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.C.); (J.C.Y.L.); (Y.S.)
| | - Joseph Caprioli
- Ophthalmology, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.C.); (J.C.Y.L.); (Y.S.)
| | - Joanne C. Y. Lee
- Ophthalmology, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.C.); (J.C.Y.L.); (Y.S.)
| | - Yifan Song
- Ophthalmology, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.C.); (J.C.Y.L.); (Y.S.)
| | - Feng-Juan Yu
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; (F.-J.Y.); (J.B.); (Y.-H.S.); (K.-K.L.); (C.-W.D.); (C.-H.T.)
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China
| | - Jingfang Bian
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; (F.-J.Y.); (J.B.); (Y.-H.S.); (K.-K.L.); (C.-W.D.); (C.-H.T.)
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China
| | - Ying-Hon Sze
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; (F.-J.Y.); (J.B.); (Y.-H.S.); (K.-K.L.); (C.-W.D.); (C.-H.T.)
| | - King-Kit Li
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; (F.-J.Y.); (J.B.); (Y.-H.S.); (K.-K.L.); (C.-W.D.); (C.-H.T.)
| | - Chi-Wai Do
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; (F.-J.Y.); (J.B.); (Y.-H.S.); (K.-K.L.); (C.-W.D.); (C.-H.T.)
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research (CEVR), The Hong Kong Polytechnic University, 17W, Hong Kong Science Park, Hong Kong, China
| | - Chi-Ho To
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; (F.-J.Y.); (J.B.); (Y.-H.S.); (K.-K.L.); (C.-W.D.); (C.-H.T.)
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research (CEVR), The Hong Kong Polytechnic University, 17W, Hong Kong Science Park, Hong Kong, China
| | - Thomas Chuen Lam
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; (F.-J.Y.); (J.B.); (Y.-H.S.); (K.-K.L.); (C.-W.D.); (C.-H.T.)
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research (CEVR), The Hong Kong Polytechnic University, 17W, Hong Kong Science Park, Hong Kong, China
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518052, China
| |
Collapse
|
11
|
Rodríguez-Solana P, Arruti N, Nieves-Moreno M, Mena R, Rodríguez-Jiménez C, Guerrero-Carretero M, Acal JC, Blasco J, Peralta JM, Del Pozo Á, Montaño VEF, Dios-Blázquez LD, Fernández-Alcalde C, González-Atienza C, Sánchez-Cazorla E, Gómez-Cano MDLÁ, Delgado-Mora L, Noval S, Vallespín E. Whole Exome Sequencing of 20 Spanish Families: Candidate Genes for Non-Syndromic Pediatric Cataracts. Int J Mol Sci 2023; 24:11429. [PMID: 37511188 PMCID: PMC10380485 DOI: 10.3390/ijms241411429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Non-syndromic pediatric cataracts are defined as opacification of the crystalline lens that occurs during the first years of life without affecting other organs. Given that this disease is one of the most frequent causes of reversible blindness in childhood, the main objective of this study was to propose new responsible gene candidates that would allow a more targeted genetic approach and expand our genetic knowledge about the disease. We present a whole exome sequencing (WES) study of 20 Spanish families with non-syndromic pediatric cataracts and a previous negative result on an ophthalmology next-generation sequencing panel. After ophthalmological evaluation and collection of peripheral blood samples from these families, WES was performed. We were able to reach a genetic diagnosis in 10% of the families analyzed and found genes that could cause pediatric cataracts in 35% of the cohort. Of the variants found, 18.2% were classified as pathogenic, 9% as likely pathogenic, and 72.8% as variants of uncertain significance. However, we did not find conclusive results in 55% of the families studied, which suggests further studies are needed. The results of this WES study allow us to propose LONP1, ACACA, TRPM1, CLIC5, HSPE1, ODF1, PIKFYVE, and CHMP4A as potential candidates to further investigate for their role in pediatric cataracts, and AQP5 and locus 2q37 as causal genes.
Collapse
Affiliation(s)
- Patricia Rodríguez-Solana
- Molecular Ophthalmology Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (P.R.-S.); (R.M.); (C.R.-J.); (V.E.F.M.); (C.G.-A.); (E.S.-C.)
| | - Natalia Arruti
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
- European Reference Network on Eye Diseases (ERN-EYE), La Paz University Hospital, 28046 Madrid, Spain
| | - María Nieves-Moreno
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
- European Reference Network on Eye Diseases (ERN-EYE), La Paz University Hospital, 28046 Madrid, Spain
| | - Rocío Mena
- Molecular Ophthalmology Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (P.R.-S.); (R.M.); (C.R.-J.); (V.E.F.M.); (C.G.-A.); (E.S.-C.)
- Biomedical Research Center in the Rare Diseases Network (CIBERER), Carlos II Health Institute (ISCIII), 28029 Madrid, Spain; (Á.D.P.); (M.d.L.Á.G.-C.); (L.D.-M.)
| | - Carmen Rodríguez-Jiménez
- Molecular Ophthalmology Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (P.R.-S.); (R.M.); (C.R.-J.); (V.E.F.M.); (C.G.-A.); (E.S.-C.)
| | - Marta Guerrero-Carretero
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
| | - Juan Carlos Acal
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
| | - Joana Blasco
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
| | - Jesús M. Peralta
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
| | - Ángela Del Pozo
- Biomedical Research Center in the Rare Diseases Network (CIBERER), Carlos II Health Institute (ISCIII), 28029 Madrid, Spain; (Á.D.P.); (M.d.L.Á.G.-C.); (L.D.-M.)
- Clinical Bioinformatics Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, CIBERER, La Paz University Hospital, 28046 Madrid, Spain;
| | - Victoria E. F. Montaño
- Molecular Ophthalmology Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (P.R.-S.); (R.M.); (C.R.-J.); (V.E.F.M.); (C.G.-A.); (E.S.-C.)
- Biomedical Research Center in the Rare Diseases Network (CIBERER), Carlos II Health Institute (ISCIII), 28029 Madrid, Spain; (Á.D.P.); (M.d.L.Á.G.-C.); (L.D.-M.)
| | - Lucía De Dios-Blázquez
- Clinical Bioinformatics Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, CIBERER, La Paz University Hospital, 28046 Madrid, Spain;
| | - Celia Fernández-Alcalde
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
| | - Carmen González-Atienza
- Molecular Ophthalmology Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (P.R.-S.); (R.M.); (C.R.-J.); (V.E.F.M.); (C.G.-A.); (E.S.-C.)
| | - Eloísa Sánchez-Cazorla
- Molecular Ophthalmology Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (P.R.-S.); (R.M.); (C.R.-J.); (V.E.F.M.); (C.G.-A.); (E.S.-C.)
| | - María de Los Ángeles Gómez-Cano
- Biomedical Research Center in the Rare Diseases Network (CIBERER), Carlos II Health Institute (ISCIII), 28029 Madrid, Spain; (Á.D.P.); (M.d.L.Á.G.-C.); (L.D.-M.)
- Clinical Genetics Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, CIBERER, La Paz University Hospital, 28046 Madrid, Spain
| | - Luna Delgado-Mora
- Biomedical Research Center in the Rare Diseases Network (CIBERER), Carlos II Health Institute (ISCIII), 28029 Madrid, Spain; (Á.D.P.); (M.d.L.Á.G.-C.); (L.D.-M.)
- Clinical Genetics Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, CIBERER, La Paz University Hospital, 28046 Madrid, Spain
| | - Susana Noval
- Department of Pediatric Ophthalmology, IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (N.A.); (M.N.-M.); (M.G.-C.); (J.C.A.); (J.B.); (J.M.P.); (C.F.-A.); (S.N.)
- European Reference Network on Eye Diseases (ERN-EYE), La Paz University Hospital, 28046 Madrid, Spain
| | - Elena Vallespín
- Molecular Ophthalmology Section, Institute of Medical and Molecular Genetics (INGEMM), IdiPaz, La Paz University Hospital, 28046 Madrid, Spain; (P.R.-S.); (R.M.); (C.R.-J.); (V.E.F.M.); (C.G.-A.); (E.S.-C.)
- European Reference Network on Eye Diseases (ERN-EYE), La Paz University Hospital, 28046 Madrid, Spain
- Biomedical Research Center in the Rare Diseases Network (CIBERER), Carlos II Health Institute (ISCIII), 28029 Madrid, Spain; (Á.D.P.); (M.d.L.Á.G.-C.); (L.D.-M.)
| |
Collapse
|
12
|
McGeoghan F, Camera E, Maiellaro M, Menon M, Huang M, Dewan P, Ziaj S, Caley MP, Donaldson M, Enright AJ, O’Toole EA. RNA sequencing and lipidomics uncovers novel pathomechanisms in recessive X-linked ichthyosis. Front Mol Biosci 2023; 10:1176802. [PMID: 37363400 PMCID: PMC10285781 DOI: 10.3389/fmolb.2023.1176802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Recessive X-linked ichthyosis (RXLI), a genetic disorder caused by deletion or point mutations of the steroid sulfatase (STS) gene, is the second most common form of ichthyosis. It is a disorder of keratinocyte cholesterol sulfate retention and the mechanism of extracutaneous phenotypes such as corneal opacities and attention deficit hyperactivity disorder are poorly understood. To understand the pathomechanisms of RXLI, the transcriptome of differentiated primary keratinocytes with STS knockdown was sequenced. The results were validated in a stable knockdown model of STS, to confirm STS specificity, and in RXLI skin. The results show that there was significantly reduced expression of genes related to epidermal differentiation and lipid metabolism, including ceramide and sphingolipid synthesis. In addition, there was significant downregulation of aldehyde dehydrogenase family members and the oxytocin receptor which have been linked to corneal transparency and behavioural disorders respectively, both of which are extracutaneous phenotypes of RXLI. These data provide a greater understanding of the causative mechanisms of RXLI's cutaneous phenotype, and show that the keratinocyte transcriptome and lipidomics can give novel insights into the phenotype of patients with RXLI.
Collapse
Affiliation(s)
- Farrell McGeoghan
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Emanuela Camera
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute-IRCCS, Rome, Italy
| | - Miriam Maiellaro
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute-IRCCS, Rome, Italy
| | - Manasi Menon
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mei Huang
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Priya Dewan
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Stela Ziaj
- Department of Dermatology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Matthew P. Caley
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Anton J. Enright
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Edel A. O’Toole
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Dermatology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
13
|
Guo X, Li C, Wang Y, Jiang C, Yang L. Long non-coding RNA nuclear paraspeckle assembly transcript 1 downregulation protects lens epithelial cells from oxidative stress-induced apoptosis by regulating the microRNA-124-3p/death-associated protein kinase 1 axis in age-related cataract. Int Ophthalmol 2023:10.1007/s10792-023-02749-4. [PMID: 37191928 DOI: 10.1007/s10792-023-02749-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/06/2023] [Indexed: 05/17/2023]
Abstract
Oxidative stress plays a significant role in cataract development. It causes the apoptosis of lens epithelial cells (LECs), resulting in lens opacification and accelerating cataract progression. Long non-coding RNAs (lncRNAs) and microRNAs have been linked to cataract development. Notably, lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) is involved in LEC apoptosis and cataract formation. However, the molecular mechanism by which NEAT1 causes age-related cataracts remains unknown. In this study, LECs (SRA01/04) were exposed to 200 μM H2O2 to generate an in vitro cataract model. The apoptosis and viability of cells were determined using flow cytometry and 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assays, respectively. Additionally, western blotting and quantitative polymerase chain reaction were used to determine the miRNA and lncRNA expression levels. When LECs were treated with hydrogen peroxide, lncRNA NEAT1 expression levels were significantly upregulated, which contributed to LEC apoptosis. Notably, lncRNA NEAT1 suppressed the expression of miR-124-3p, a critical regulator of apoptosis, whereas NEAT1 inhibition increased miR-124-3p expression and alleviated apoptosis. However, this effect was reversed when miR1243p expression was inhibited. Additionally, the miR1243p mimic effectively inhibited the death-associated protein kinase 1 (DAPK1) expression and apoptosis of LECs, while the DAPK1 mimic reversed these effects. In conclusion, our findings indicate that the lncRNA NEAT1/miR-124-3p/DAPK1 signaling loop is involved in the regulation of LEC apoptosis induced by oxidative stress, which can be exploited to develop potential treatment strategies for age-related cataracts.
Collapse
Affiliation(s)
- Xuanni Guo
- Department of Ophthalmology, Xianyang Central Hospital, No.78 Renmin East Road, Xianyang, 712000, China
| | - Chunyan Li
- Department of Ophthalmology, Xianyang Central Hospital, No.78 Renmin East Road, Xianyang, 712000, China.
| | - Yongbin Wang
- Department of Ophthalmology, Xianyang Central Hospital, No.78 Renmin East Road, Xianyang, 712000, China
| | - Chunhui Jiang
- Department of Ophthalmology, Xianyang Central Hospital, No.78 Renmin East Road, Xianyang, 712000, China
| | - Li Yang
- Department of Ophthalmology, Xianyang Central Hospital, No.78 Renmin East Road, Xianyang, 712000, China
| |
Collapse
|
14
|
Voulgaridou GP, Theologidis V, Venetikidou M, Tsochantaridis I, Tsolou A, Koffa M, Panayiotidis MI, Pappa A. Investigating the Functional Roles of Aldehyde Dehydrogenase 3A1 in Human Corneal Epithelial Cells. Int J Mol Sci 2023; 24:ijms24065845. [PMID: 36982917 PMCID: PMC10056195 DOI: 10.3390/ijms24065845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Aldehyde dehydrogenase 3A1 (ALDH3A1) oxidizes medium-chain aldehydes to their corresponding carboxylic acids. It is expressed at high rates in the human cornea, where it has been characterized as a multi-functional protein displaying various cytoprotective modes of action. Previous studies identified its association with the DNA damage response (DDR) pathway. Here, we utilized a stable transfected HCE-2 (human corneal epithelium) cell line expressing ALDH3A1, to investigate the molecular mechanisms underlying the cytoprotective role(s) of ALDH3A1. Our data revealed morphological differences among the ALDH3A1-expressing and the mock-transfected HCE-2 cells accompanied by differential expression of E-cadherin. Similarly, the ALDH3A1/HCE-2 cells demonstrated higher mobility, reduced proliferation, upregulation of ZEB1, and downregulation of CDK3, and p57. The expression of ALDH3A1 also affected cell cycle progression by inducing the sequestration of HCE-2 cells at the G2/M phase. Following 16 h cell treatments with either H2O2 or etoposide, a significantly lower percentage of ALDH3A1/HCE-2 cells were apoptotic compared to the respective treated mock/HCE-2 cells. Interestingly, the protective effect of ALDH3A1 expression under these oxidative and genotoxic conditions was accompanied by a reduced formation of γ-H2AX foci and higher levels of total and phospho (Ser15) p53. Finally, ALDH3A1 was found to be localized both in the cytoplasm and the nucleus of transfected HCE-2 cells. Its cellular compartmentalization was not affected by oxidant treatment, while the mechanism by which ALDH3A1 translocates to the nucleus remains unknown. In conclusion, ALDH3A1 protects cells from both apoptosis and DNA damage by interacting with key homeostatic mechanisms associated with cellular morphology, cell cycle, and DDR.
Collapse
Affiliation(s)
- Georgia-Persephoni Voulgaridou
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Vasileios Theologidis
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Maria Venetikidou
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Ilias Tsochantaridis
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Avgi Tsolou
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Maria Koffa
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Mihalis I Panayiotidis
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Ayios Dometios, Nicosia 2371, Cyprus
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
15
|
Wang S, Sun M, Ning Z, Chen Y, Zhou H, Mu W. The effects of sustained and diel-cycling hypoxia on high-latitude fish Phoxinus lagowskii. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 45:101059. [PMID: 36706598 DOI: 10.1016/j.cbd.2023.101059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/21/2023]
Abstract
High-latitude fish are subjected to sustained and diel-cycling hypoxia. Oxygen deficiency could pose a serious threat to fish, but little information is available regarding the response mechanisms employed by high-latitude fish to sustained and diel-cycling hypoxia. In this study, a combination of transcriptomics and metabolomics were used to examine the molecular response mechanisms actioned by sustained and diel-cycling hypoxia in the high-latitude fish, Phoxinus lagowskii. P. lagowskii was divided into normoxic control (6.0-7.0 mg/L dissolved oxygen), sustained (1.5 mg/L dissolved oxygen), and diel-cycling hypoxic treatment (6.0-7.0 mg/L between 07:00-21:00, and 3.0-4.0 mg/L between 21:00-07:00) tanks for 28 days. Differentially expressed genes (DEGs) and significantly different metabolites (DMs) related to digestive proteases, lipid metabolism, estrogen signaling pathway, steroid hormone biosynthesis, glutathione metabolism, and tryptophan metabolism were identified from comparative metabolomic and transcriptomic data expression profiles within the liver. The current study found that P. lagowskii had significantly different responses between sustained and diel-cycling hypoxia. P. lagowskii faced with sustained hypoxia may enhance their tolerance capacity through phospholipid and glutathione metabolism. Our data provide new insights into the high latitude fish coping with changes in hypoxia and warrants further investigation into these potentially important genes and metabolites.
Collapse
Affiliation(s)
- Sihan Wang
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Mingyang Sun
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Zhaoyang Ning
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Yingqiao Chen
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Haishui Zhou
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Weijie Mu
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China.
| |
Collapse
|
16
|
Beutler M, Harnischfeger J, Weber MHW, Hahnel SR, Quack T, Blohm A, Ueberall ME, Timm T, Lochnit G, Rennar GA, Gallinger TL, Houhou H, Rahlfs S, Falcone FH, Becker K, Schlitzer M, Haeberlein S, Czermak P, Salzig D, Grevelding CG. Identification and characterisation of the tegument-expressed aldehyde dehydrogenase SmALDH_312 of Schistosoma mansoni, a target of disulfiram. Eur J Med Chem 2023; 251:115179. [PMID: 36948075 DOI: 10.1016/j.ejmech.2023.115179] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
Schistosomiasis is an infectious disease caused by blood flukes of the genus Schistosoma and affects approximately 200 million people worldwide. Since Praziquantel (PZQ) is the only drug for schistosomiasis, alternatives are needed. By a biochemical approach, we identified a tegumentally expressed aldehyde dehydrogenase (ALDH) of S. mansoni, SmALDH_312. Molecular analyses of adult parasites showed Smaldh_312 transcripts in both genders and different tissues. Physiological and cell-biological experiments exhibited detrimental effects of the drug disulfiram (DSF), a known ALDH inhibitor, on larval and adult schistosomes in vitro. DSF also reduced stem-cell proliferation and caused severe tegument damage in treated worms. In silico-modelling of SmALDH_312 and docking analyses predicted DSF binding, which we finally confirmed by enzyme assays with recombinant SmALDH_312. Furthermore, we identified compounds of the Medicine for Malaria Venture (MMV) pathogen box inhibiting SmALDH_312 activity. Our findings represent a promising starting point for further development towards new drugs for schistosomiasis.
Collapse
Affiliation(s)
- Mandy Beutler
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Julie Harnischfeger
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Michael H W Weber
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Steffen R Hahnel
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Thomas Quack
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Ariane Blohm
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Monique E Ueberall
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany; Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Thomas Timm
- Protein Analytics, Institute of Biochemistry, Justus Liebig University Giessen, Germany
| | - Günter Lochnit
- Protein Analytics, Institute of Biochemistry, Justus Liebig University Giessen, Germany
| | - Georg A Rennar
- Department of Pharmaceutical Chemistry, Philipps Universität Marburg, Germany, Germany
| | - Tom L Gallinger
- Department of Pharmaceutical Chemistry, Philipps Universität Marburg, Germany, Germany
| | - Hicham Houhou
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Stefan Rahlfs
- Institute for Biochemistry and Molecular Biology, Interdisciplinary Research Centre, Justus Liebig University, Germany
| | - Franco H Falcone
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Katja Becker
- Institute for Biochemistry and Molecular Biology, Interdisciplinary Research Centre, Justus Liebig University, Germany
| | - Martin Schlitzer
- Department of Pharmaceutical Chemistry, Philipps Universität Marburg, Germany, Germany
| | - Simone Haeberlein
- Institute of Parasitology, BFS, Justus Liebig University Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany
| | | |
Collapse
|
17
|
Xia J, Li S, Liu S, Zhang L. Aldehyde dehydrogenase in solid tumors and other diseases: Potential biomarkers and therapeutic targets. MedComm (Beijing) 2023; 4:e195. [PMID: 36694633 PMCID: PMC9842923 DOI: 10.1002/mco2.195] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 01/18/2023] Open
Abstract
The family of aldehyde dehydrogenases (ALDHs) contains 19 isozymes and is involved in the oxidation of endogenous and exogenous aldehydes to carboxylic acids, which contributes to cellular and tissue homeostasis. ALDHs play essential parts in detoxification, biosynthesis, and antioxidants, which are of important value for cell proliferation, differentiation, and survival in normal body tissues. However, ALDHs are frequently dysregulated and associated with various diseases like Alzheimer's disease, Parkinson's disease, and especially solid tumors. Notably, the involvement of the ALDHs in tumor progression is responsible for the maintenance of the stem-cell-like phenotype, triggering rapid and aggressive clinical progressions. ALDHs have captured increasing attention as biomarkers for disease diagnosis and prognosis. Nevertheless, these require further longitudinal clinical studies in large populations for broad application. This review summarizes our current knowledge regarding ALDHs as potential biomarkers in tumors and several non-tumor diseases, as well as recent advances in our understanding of the functions and underlying molecular mechanisms of ALDHs in disease development. Finally, we discuss the therapeutic potential of ALDHs in diseases, especially in tumor therapy with an emphasis on their clinical implications.
Collapse
Affiliation(s)
- Jie Xia
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Siqin Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer MedicineNanjing Medical UniversityNanjingChina
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
18
|
Chhunchha B, Kubo E, Krueger RR, Singh DP. Hydralazine Revives Cellular and Ocular Lens Health-Span by Ameliorating the Aging and Oxidative-Dependent Loss of the Nrf2-Activated Cellular Stress Response. Antioxidants (Basel) 2023; 12:140. [PMID: 36671002 PMCID: PMC9854670 DOI: 10.3390/antiox12010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/10/2023] Open
Abstract
A major hallmark of aging-associated diseases is the inability to evoke cellular defense responses. Transcriptional protein Nrf2 (nuclear factor erythroid-derived 2-related factor) plays a pivotal role in the oxidative stress response, cellular homeostasis, and health span. Nrf2's activation has been identified as a therapeutic target to restore antioxidant defense in aging. Here, we demonstrated that FDA-approved drug, hydralazine (Hyd), was a reactivator of the Nrf2/ARE (antioxidant response element) pathway in various ages and types of mouse (m) or human (h) lens epithelial cells (LECs) and mice lenses in-vitro/in-vivo. This led to Hyd-driven abatement of carbonyls, reduced reactive oxygen species (ROS), and reduced 4-HNE/MDA-adducts with cytoprotection, and extended lens healthspan by delaying/preventing lens opacity against aging/oxidative stress. We elucidated that Hyd activated the protective signaling by inducing Nrf2 to traverse from the cytoplasm to the nucleus and potentiated the ARE response by direct interaction of Nrf2 and ARE sequences of the promoter. Loss-of-function study and cotreatment of Hyd and antioxidant, N-acetyl cysteine (NAC) or Peroxiredoxin (Prdx)6, specified that Nrf2/ARE-driven increase in the promoter activity was Hyd-dependent. Our study provides proof-of concept evidence and, thereby, paves the way to repurposing Hyd as a therapeutic agent to delay/prevent aging and oxidative-related disorders.
Collapse
Affiliation(s)
- Bhavana Chhunchha
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 9200293, Japan
| | - Ronald R. Krueger
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dhirendra P. Singh
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
19
|
Voulgaridou GP, Theologidis V, Xanthis V, Papagiannaki E, Tsochantaridis I, Fadouloglou VE, Pappa A. Identification of a peptide ligand for human ALDH3A1 through peptide phage display: Prediction and characterization of protein interaction sites and inhibition of ALDH3A1 enzymatic activity. Front Mol Biosci 2023; 10:1161111. [PMID: 37021113 PMCID: PMC10067601 DOI: 10.3389/fmolb.2023.1161111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
Aldehyde dehydrogenase 3A1 (ALDH3A1) by oxidizing medium chain aldehydes to their corresponding carboxylic acids, is involved in the detoxification of toxic byproducts and is considered to play an important role in antioxidant cellular defense. ALDH3A1 has been implicated in various other functions such as cell proliferation, cell cycle regulation, and DNA damage response. Recently, it has been identified as a putative biomarker of prostate, gastric, and lung cancer stem cell phenotype. Although ALDH3A1 has multifaceted functions in both normal and cancer homeostasis, its modes of action are currently unknown. To this end, we utilized a random 12-mer peptide phage display library to identify efficiently human ALDH3A1-interacting peptides. One prevailing peptide (P1) was systematically demonstrated to interact with the protein of interest, which was further validated in vitro by peptide ELISA. Bioinformatic analysis indicated two putative P1 binding sites on the protein surface implying biomedical potential and potent inhibitory activity of the P1 peptide on hALDH3A1 activity was demonstrated by enzymatic studies. Furthermore, in search of potential hALDH3A1 interacting players, a BLASTp search demonstrated that no protein in the database includes the full-length amino acid sequence of P1, but identified a list of proteins containing parts of the P1 sequence, which may prove potential hALDH3A1 interacting partners. Among them, Protein Kinase C Binding Protein 1 and General Transcription Factor II-I are candidates of high interest due to their cellular localization and function. To conclude, this study identifies a novel peptide with potential biomedical applications and further suggests a list of protein candidates be explored as possible hALDH3A1-interacting partners in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Aglaia Pappa
- *Correspondence: Vasiliki E. Fadouloglou, ; Aglaia Pappa,
| |
Collapse
|
20
|
Pichler R, Rizzo L, Tröndle K, Bühler M, Brucker H, Müller AL, Grand K, Farè S, Viau A, Kaminski MM, Kuehn EW, Koch F, Zimmermann S, Koltay P, Lienkamp SS. Tuning the 3D microenvironment of reprogrammed tubule cells enhances biomimetic modeling of polycystic kidney disease. Biomaterials 2022; 291:121910. [DOI: 10.1016/j.biomaterials.2022.121910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022]
|
21
|
Identification of lncRNAs Associated with the Pathogenesis of Diabetic Retinopathy: From Sequencing Analysis to Validation via In Vivo and In Vitro Experiments. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1755945. [PMID: 36299680 PMCID: PMC9592201 DOI: 10.1155/2022/1755945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022]
Abstract
This study is aimed at screening for differentially expressed long noncoding RNAs (lncRNAs) associated with the pathogenesis of diabetic retinopathy and verifying the role of lncZNRD1 in high glucose-induced injury of retinal microvascular endothelial cells. The retinal tissues of normal and diabetic rats were collected for high-throughput sequencing of differentially expressed lncRNAs. Retinal microvascular endothelial cells were treated with 50 mM glucose for 4 h, 8 h, 24 h, 48 h, and 72 h. Our results showed that compared with the control group, there were 736 differentially expressed lncRNAs in the retina tissue of the model group, including 226 upregulated genes and 736 downregulated genes. Based on the differentially expressed lncRNAs, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated that the ErbB signaling pathway, transforming growth factor- (TGF-) β signaling pathway, PI3K − Akt signaling pathway, cyclic adenosine 3,5-monophosphate (cAMP) signaling pathway, mitogen-activated protein kinase (MAPK) signaling pathway, and hypoxia-inducible factor-1 (HIF-1) signaling pathway were likely involved in the regulation of diabetic retinopathy. Compared with the control group, the expression of lncZNRD1-AS1 was significantly increased in retinal microvascular endothelial cells after treatment with high glucose for 24 h. Silencing lncZNRD1 promoted high glucose-induced apoptosis of microvascular endothelial cells. Additionally, silencing lncZNRD1 increased the expression levels of ALDH7A1 and ALDH3A2. In conclusion, lncZNRD1-AS1 demonstrated potentially beneficial function against high glucose-induced retina cell injury by regulating ALDH7A1 and ALDH3A2 expressions.
Collapse
|
22
|
Zhang G, Kang L, Li P, Ran Q, Chen X, Ji M, Guan H. Genome-wide repertoire of transfer RNA-derived fragments in a mouse model of age-related cataract. Curr Eye Res 2022; 47:1397-1404. [PMID: 35930684 DOI: 10.1080/02713683.2022.2110263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Purpose To investigate the roles of tRNA-derived small RNAs (tsRNAs) containing transfer RNA-derived fragments (tRFs) and tRNA halves in age-related cataract (ARC). Methods: Lens capsule tissue from Emory mice at 3 months and 8 months of age were dissected for integrated tsRNA and gene transcriptome sequencing. Quantitative real-time PCR assay (qRT-PCR) was perform for validating sequencing results. Bioinformatics analysis was constructed to reveal the roles of tsRNAs. Results: A total of 422 DE tsRNAs were changed, in which 156 were elevated while 266 were declined in 8-month-old mice. Subsequently, the gene sequencing data exhibited 375 upregulated and 456 downregulated DE genes. Validation by qRT-PCR in 5 selected upregulated tRFs was consistent with tsRNAs sequencing results. Moreover, bioinformatics analysis identified 25 downregulated target genes of the 5 validated tRFs. Furthermore, GO analysis revealed that these target genes were mainly enriched in camera-type eye development, sensory organ development and so on. Conclusion: Our study provide a novel perspective for the role of tsRNAs in pathogenesis of ARC, and thus therapeutic potential targets for ARC.
Collapse
Affiliation(s)
- Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Lihua Kang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Pengfei Li
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Qiliang Ran
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiang Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Min Ji
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
23
|
Berdyński M, Krawczyk P, Safranow K, Borzemska B, Szaflik JP, Nowakowska-Żawrocka K, Żekanowski C, Giebułtowicz J. Common ALDH3A1 Gene Variant Associated with Keratoconus Risk in the Polish Population. J Clin Med 2021; 11:jcm11010008. [PMID: 35011749 PMCID: PMC8745142 DOI: 10.3390/jcm11010008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
Background: ALDH3A1 protein is important in maintaining corneal physiology and protecting the eye from UV damage. However, none of the genome-wide association studies has indicated that the ALDH3A1 locus is associated with keratoconus. In this study, we examined the potential role of ALDH3A1 variants as risk factors for keratoconus incidence and severity in a large group of Polish keratoconus patients. Methods: In the first stage we analyzed the coding region sequence of the ALDH3A1 in a subgroup of keratoconus. Then, we genotyped three selected ALDH3A1 variants in a larger KC group of patients (n = 261) and healthy controls (n = 317). Results: We found that the rs1042183 minor allele A is a risk factor for keratoconus in the dominant model (OR = 2.06, 95%CI = 1.42–2.98, p = 0.00013). The rs2228100 variant genotypes appear to be associated with an earlier age of KC diagnosis in the Polish population (p = 0.055 for comparison of three genotypes and p = 0.022 for the dominant inheritance model). Conclusions: The rs1042183 variant in ALDH3A1 is associated with keratoconus risk in the Polish population. The differences in the allele frequency between both populations could be partially responsible for the difference in the disease prevalence.
Collapse
Affiliation(s)
- Mariusz Berdyński
- Laboratory of Neurogenetics, Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland; (M.B.); (B.B.); (C.Ż.)
| | - Piotr Krawczyk
- Department of Ophthalmology, Medical University of Warsaw, 13 Sierakowskiego Str., 03-709 Warsaw, Poland; (P.K.); (J.P.S.)
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 72 Powstańców Wlkp. Str., 70-111 Szczecin, Poland;
| | - Beata Borzemska
- Laboratory of Neurogenetics, Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland; (M.B.); (B.B.); (C.Ż.)
| | - Jacek P. Szaflik
- Department of Ophthalmology, Medical University of Warsaw, 13 Sierakowskiego Str., 03-709 Warsaw, Poland; (P.K.); (J.P.S.)
| | - Karolina Nowakowska-Żawrocka
- Department of Bioanalysis and Drugs Analysis, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland;
| | - Cezary Żekanowski
- Laboratory of Neurogenetics, Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland; (M.B.); (B.B.); (C.Ż.)
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drugs Analysis, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland;
- Correspondence:
| |
Collapse
|
24
|
Thompson B, Chen Y, Davidson EA, Garcia-Milian R, Golla JP, Apostolopoulos N, Orlicky DJ, Schey K, Thompson DC, Vasiliou V. Impaired GSH biosynthesis disrupts eye development, lens morphogenesis and PAX6 function. Ocul Surf 2021; 22:190-203. [PMID: 34425299 DOI: 10.1016/j.jtos.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/29/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE The purpose of this study was to elucidate the role and molecular consequences of impaired glutathione (GSH) biosynthesis on eye development. METHODS GSH biosynthesis was impaired in surface ectoderm-derived ocular tissues by crossing Gclcf/f mice with hemizygous Le-Cre transgenic mice to produce Gclcf/f/Le-CreTg/- (KO) mice. Control mice included Gclcf/fand Gclcwt/wt/Le-CreTg/- mice (CRE). Eyes from all mice (at various stages of eye development) were subjected to histological, immunohistochemical, Western blot, RT-qPCR, RNA-seq, and subsequent Gene Ontology, Ingenuity Pathway Analysis and TRANSFAC analyses. PAX6 transactivation activity was studied using a luciferase reporter assay in HEK293T cells depleted of GSH using buthionine sulfoximine (BSO). RESULTS Deletion of Gclc diminished GSH levels, increased reactive oxygen species (ROS), and caused an overt microphthalmia phenotype characterized by malformation of the cornea, iris, lens, and retina that is distinct from and much more profound than the one observed in CRE mice. In addition, only the lenses of KO mice displayed reduced crystallin (α, β), PITX3 and Foxe3 expression. RNA-seq analyses at postnatal day 1 revealed 1552 differentially expressed genes (DEGs) in the lenses of KO mice relative to those from Gclcf/f mice, with Crystallin and lens fiber cell identity genes being downregulated while lens epithelial cell identity and immune response genes were upregulated. Bioinformatic analysis of the DEGs implicated PAX6 as a key upstream regulator. PAX6 transactivation activity was impaired in BSO-treated HEK293T cells. CONCLUSIONS These data suggest that impaired ocular GSH biosynthesis may disrupt eye development and PAX6 function.
Collapse
Affiliation(s)
- Brian Thompson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA
| | - Emily A Davidson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, USA
| | - Jaya Prakash Golla
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA; Department of Medicine, Yale University School of Medicine, New Haven, CT, USA; Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | | | - David J Orlicky
- Department of Pathology, Anschutz School of Medicine, University of Colorado, Aurora, CO, USA
| | - Kevin Schey
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David C Thompson
- Department of Clinical Pharmacy, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, USA.
| |
Collapse
|
25
|
Faranda AP, Shihan MH, Wang Y, Duncan MK. The effect of sex on the mouse lens transcriptome. Exp Eye Res 2021; 209:108676. [PMID: 34146586 DOI: 10.1016/j.exer.2021.108676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The transcriptome of mammalian tissues differs between males and females, and these differences can change across the lifespan, likely regulating known sexual dimorphisms in disease prevalence and severity. Cataract, the most prevalent disease of the ocular lens, occurs at similar rates in young individuals, but its incidence is elevated in older women compared to men of the same age. However, the influence of sex on the lens transcriptome was unknown. RNAseq based transcriptomic profiling of young adult C57BL/6J mouse lens epithelial and fiber cells revealed that few genes are differentially expressed between the sexes. In contrast, lens cells from aged (24 month old) male and female C57BL/6J mice differentially expressed many genes, including several whose expression is lens preferred. Like cataracts, posterior capsular opacification (PCO), a major sequela of cataract surgery, may also be more prevalent in women. Lens epithelial cells isolated from mouse eyes 24 h after lens fiber cell removal exhibited numerous transcriptomic differences between the sexes, including genes implicated in complement cascades and extracellular matrix regulation, and these differences are much more pronounced in aged mice than in young mice. These results provide an unbiased basis for future studies on how sex affects the lens response to aging, cataract development, and cataract surgery.
Collapse
Affiliation(s)
- Adam P Faranda
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA
| | - Mahbubul H Shihan
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA
| | - Yan Wang
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA
| | - Melinda K Duncan
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
26
|
Dysfunction of the limbal epithelial stem cell niche in aniridia-associated keratopathy. Ocul Surf 2021; 21:160-173. [PMID: 34102310 DOI: 10.1016/j.jtos.2021.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/22/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE Abnormalities in the limbal niche microenvironment have been suggested to be causally involved in aniridia-associated keratopathy (AAK), but histological analyses on the limbal structure and composition in AAK are lacking. Here, we investigated morphologic and molecular alterations of the limbal epithelial stem cell niche in human congenital aniridia. METHODS The blind, buphthalmic and painful left eye of a 16-year old girl with congenital aniridia and juvenile glaucoma had to be enucleated because of uncontrolled intraocular pressure. The diagnosis of AAK was based on classical clinical features and partial limbal stem cell deficiency in the superior half. Genetic analysis identified a large heterozygous PAX6 gene deletion encompassing exons 11-15 as well as exon 9 of the neighboring ELP4 gene. Three limbal biopsies were taken from the superior, nasal and temporal regions to isolate and cultivate limbal epithelial progenitor cells and subject them to mRNA expression analyses. The globe was vertically bisected and processed for light and transmission electron microscopy and immunohistochemistry. RESULTS Comparative analysis of the superior and inferior limbal zones showed a gradual degradation of palisade structures associated with the transition from a hyperplastic to an attenuated corneal epithelium, inflammatory cell infiltrations and basement membrane irregularities. The clinically unaffected inferior part revealed no distinct stem cell clusters in the preserved palisade region, but a uniform population of hyperproliferative, undifferentiated progenitor cells in the basal/suprabasal layers of limbal and corneal epithelia, which gave rise to maldifferentiated epithelial cells exhibiting a conjunctival/epidermal phenotype and nuclear-to-cytoplasmic translocation of Pax6. The structure of the limbal niche was fundamentally perturbed, showing marked alterations in extracellular matrix composition, dislocation of atypical melanocytes lacking melanosomes and melanin, aberrant Wnt/β-catenin and retinoic acid signaling, and massive immune cell infiltration. CONCLUSIONS Considering the limitations of a single Case study, the findings suggest that ocular surface alterations in AAK are caused by a primary dysfunction and gradual breakdown of the limbal stem cell niche through Pax6-related effects on both melanogenesis and epithelial differentiation.
Collapse
|
27
|
Yu Q, Biswas S, Ma G, Zhao P, Li B, Li J. Canonical NF-κB signaling maintains corneal epithelial integrity and prevents corneal aging via retinoic acid. eLife 2021; 10:e67315. [PMID: 34085926 PMCID: PMC8192125 DOI: 10.7554/elife.67315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
Disorders of the transparent cornea affect millions of people worldwide. However, how to maintain and/or regenerate this organ remains unclear. Here, we show that Rela (encoding a canonical NF-κB subunit) ablation in K14+ corneal epithelial stem cells not only disrupts corneal regeneration but also results in age-dependent epithelial deterioration, which triggers aberrant wound-healing processes including stromal remodeling, neovascularization, epithelial metaplasia, and plaque formation at the central cornea. These anomalies are largely recapitulated in normal mice that age naturally. Mechanistically, Rela deletion suppresses expression of Aldh1a1, an enzyme required for retinoic acid synthesis from vitamin A. Retinoic acid administration blocks development of ocular anomalies in Krt14-Cre; Relaf/f mice and naturally aged mice. Moreover, epithelial metaplasia and plaque formation are preventable by inhibition of angiogenesis. This study thus uncovers the major mechanisms governing corneal maintenance, regeneration, and aging and identifies the NF-κB-retinoic acid pathway as a therapeutic target for corneal disorders.
Collapse
MESH Headings
- Age Factors
- Aldehyde Dehydrogenase 1 Family/genetics
- Aldehyde Dehydrogenase 1 Family/metabolism
- Animals
- Burns, Chemical/drug therapy
- Burns, Chemical/etiology
- Burns, Chemical/metabolism
- Burns, Chemical/pathology
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cellular Senescence/drug effects
- Corneal Neovascularization/metabolism
- Corneal Neovascularization/pathology
- Corneal Neovascularization/prevention & control
- Corneal Stroma/drug effects
- Corneal Stroma/metabolism
- Corneal Stroma/pathology
- Disease Models, Animal
- Epithelium, Corneal/drug effects
- Epithelium, Corneal/metabolism
- Epithelium, Corneal/pathology
- Eye Burns/chemically induced
- Eye Burns/drug therapy
- Eye Burns/metabolism
- Eye Burns/pathology
- Mice, Knockout
- Regeneration/drug effects
- Retinal Dehydrogenase/genetics
- Retinal Dehydrogenase/metabolism
- Signal Transduction
- Stem Cells/drug effects
- Stem Cells/metabolism
- Stem Cells/pathology
- Transcription Factor RelA/genetics
- Transcription Factor RelA/metabolism
- Tretinoin/pharmacology
- Mice
Collapse
Affiliation(s)
- Qian Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghaiChina
| | - Soma Biswas
- Department of Ophthalmology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Gang Ma
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghaiChina
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghaiChina
- Institute of Traditional Chinese Medicine and Stem Cell Research, School of Basic Medicine, Chengdu University of Traditional Chinese MedicineChengduChina
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
28
|
Shortall K, Djeghader A, Magner E, Soulimane T. Insights into Aldehyde Dehydrogenase Enzymes: A Structural Perspective. Front Mol Biosci 2021; 8:659550. [PMID: 34055881 PMCID: PMC8160307 DOI: 10.3389/fmolb.2021.659550] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022] Open
Abstract
Aldehyde dehydrogenases engage in many cellular functions, however their dysfunction resulting in accumulation of their substrates can be cytotoxic. ALDHs are responsible for the NAD(P)-dependent oxidation of aldehydes to carboxylic acids, participating in detoxification, biosynthesis, antioxidant and regulatory functions. Severe diseases, including alcohol intolerance, cancer, cardiovascular and neurological diseases, were linked to dysfunctional ALDH enzymes, relating back to key enzyme structure. An in-depth understanding of the ALDH structure-function relationship and mechanism of action is key to the understanding of associated diseases. Principal structural features 1) cofactor binding domain, 2) active site and 3) oligomerization mechanism proved critical in maintaining ALDH normal activity. Emerging research based on the combination of structural, functional and biophysical studies of bacterial and eukaryotic ALDHs contributed to the appreciation of diversity within the superfamily. Herewith, we discuss these studies and provide our interpretation for a global understanding of ALDH structure and its purpose–including correct function and role in disease. Our analysis provides a synopsis of a common structure-function relationship to bridge the gap between the highly studied human ALDHs and lesser so prokaryotic models.
Collapse
Affiliation(s)
- Kim Shortall
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, Ireland
| | - Ahmed Djeghader
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, Ireland
| | - Edmond Magner
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, Ireland
| | - Tewfik Soulimane
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
29
|
Verma M, Khan MIK, Kadumuri RV, Chakrapani B, Awasthi S, Mahesh A, Govindaraju G, Chavali PL, Rajavelu A, Chavali S, Dhayalan A. PRMT3 interacts with ALDH1A1 and regulates gene-expression by inhibiting retinoic acid signaling. Commun Biol 2021; 4:109. [PMID: 33495566 PMCID: PMC7835222 DOI: 10.1038/s42003-020-01644-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 12/15/2020] [Indexed: 12/23/2022] Open
Abstract
Protein arginine methyltransferase 3 (PRMT3) regulates protein functions by introducing asymmetric dimethylation marks at the arginine residues in proteins. However, very little is known about the interaction partners of PRMT3 and their functional outcomes. Using yeast-two hybrid screening, we identified Retinal dehydrogenase 1 (ALDH1A1) as a potential interaction partner of PRMT3 and confirmed this interaction using different methods. ALDH1A1 regulates variety of cellular processes by catalyzing the conversion of retinaldehyde to retinoic acid. By molecular docking and site-directed mutagenesis, we identified the specific residues in the catalytic domain of PRMT3 that facilitate interaction with the C-terminal region of ALDH1A1. PRMT3 inhibits the enzymatic activity of ALDH1A1 and negatively regulates the expression of retinoic acid responsive genes in a methyltransferase activity independent manner. Our findings show that in addition to regulating protein functions by introducing methylation modifications, PRMT3 could also regulate global gene expression through protein-protein interactions. Here, the authors demonstrate that protein arginine methyltransferase 3 (PRMT3) interacts with and inhibits the retinal dehydrogenase ALDH1A1, negatively regulating the expression of retinoic acid responsive genes. This study shows that PRMT3 affects diverse biological processes not only by globally regulating protein function through methylation but also by regulating gene expression.
Collapse
Affiliation(s)
- Mamta Verma
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Mohd Imran K Khan
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Rajashekar Varma Kadumuri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, 517507, India
| | - Baskar Chakrapani
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Sharad Awasthi
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Arun Mahesh
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India
| | - Gayathri Govindaraju
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Pavithra L Chavali
- CSIR-Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Arumugam Rajavelu
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Sreenivas Chavali
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, 517507, India.
| | - Arunkumar Dhayalan
- Department of Biotechnology, Pondicherry University, Puducherry, 605014, India.
| |
Collapse
|
30
|
Abstract
Drug metabolizing enzymes catalyze the biotransformation of many of drugs and chemicals. The drug metabolizing enzymes are distributed among several evolutionary families and catalyze a range of detoxication reactions, including oxidation/reduction, conjugative, and hydrolytic reactions that serve to detoxify potentially toxic compounds. This detoxication function requires that drug metabolizing enzymes exhibit substrate promiscuity. In addition to their catalytic functions, many drug metabolizing enzymes possess functions unrelated to or in addition to catalysis. Such proteins are termed 'moonlighting proteins' and are defined as proteins with multiple biochemical or biophysical functions that reside in a single protein. This review discusses the diverse moonlighting functions of drug metabolizing enzymes and the roles they play in physiological functions relating to reproduction, vision, cell signaling, cancer, and transport. Further research will likely reveal new examples of moonlighting functions of drug metabolizing enzymes.
Collapse
Affiliation(s)
- Philip G Board
- John Curtin School of Medical Research, ANU College of Health and Medicine, The Australian National University, Canberra, ACT, Australia
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, New York, NY, USA
| |
Collapse
|
31
|
Srivastava O, Srivastava K, Joseph R, Wilson L. Increased Association of Deamidated αA- N101D with Lens membrane of transgenic αA N101D vs. wild type αA mice: potential effects on intracellular ionic imbalance and membrane disorganization. BMC Ophthalmol 2020; 20:484. [PMID: 33302904 PMCID: PMC7726915 DOI: 10.1186/s12886-020-01734-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/13/2020] [Indexed: 11/10/2022] Open
Abstract
We have generated two mouse models, in one by inserting the human lens αAN101D transgene in CRYαAN101D mice, and in the other by inserting human wild-type αA-transgene in CRYαAWT mice. The CRYαAN101D mice developed cortical cataract at about 7-months of age relative to CRYαAWT mice. The objective of the study was to determine the following relative changes in the lenses of CRYαAN101D- vs. CRYαAWT mice: age-related changes with specific emphasis on protein insolubilization, relative membrane-association of αAN101D vs. WTαA proteins, and changes in intracellular ionic imbalance and membrane organization. METHODS Lenses of varying ages from CRYαAWT and CRYαAN101D mice were compared for an age-related protein insolubilization. The relative lens membrane-association of the αAN101D- and WTαA proteins in the two types of mice was determined by immunohistochemical-, immunogold-labeling-, and western blot analyses. The relative levels of membrane-binding of recombinant αAN101D- and WTαA proteins was determined by an in vitro assay, and the levels of intracellular Ca2+ uptake and Na, K-ATPase mRNA were determined in the cultured epithelial cells from lenses of the two types of mice. RESULTS Compared to the lenses of CRYαAWT, the lenses of CRYαAN101D mice exhibited: (A) An increase in age-related protein insolubilization beginning at about 4-months of age. (B) A greater lens membrane-association of αAN101D- relative to WTαA protein during immunogold-labeling- and western blot analyses, including relatively a greater membrane swelling in the CRYαAN101D lenses. (C) During in vitro assay, the greater levels of binding αAN101D- relative to WTαA protein to membranes was observed. (D) The 75% lower level of Na, K-ATPase mRNA but 1.5X greater Ca2+ uptake were observed in cultured lens epithelial cells of CRYαAN101D- than those of CRYαAWT mice. CONCLUSIONS The results show that an increased lens membrane association of αAN101D--relative WTαA protein in CRYαAN101D mice than CRYαAWT mice occurs, which causes intracellular ionic imbalance, and in turn, membrane swelling that potentially leads to cortical opacity.
Collapse
Affiliation(s)
- Om Srivastava
- Department of Optometry and Vision Science, University of Alabama at Birmingham, 1716, University Boulevard, Birmingham, AL, 35294-0010, USA.
| | - Kiran Srivastava
- Department of Optometry and Vision Science, University of Alabama at Birmingham, 1716, University Boulevard, Birmingham, AL, 35294-0010, USA
| | - Roy Joseph
- Department of Optometry and Vision Science, University of Alabama at Birmingham, 1716, University Boulevard, Birmingham, AL, 35294-0010, USA
| | - Landon Wilson
- Targeted Metabolomics and Proteomics Laboratory (TMPL), Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294-0010, USA
| |
Collapse
|
32
|
Lou B, Boger M, Bennewitz K, Sticht C, Kopf S, Morgenstern J, Fleming T, Hell R, Yuan Z, Nawroth PP, Kroll J. Elevated 4-hydroxynonenal induces hyperglycaemia via Aldh3a1 loss in zebrafish and associates with diabetes progression in humans. Redox Biol 2020; 37:101723. [PMID: 32980661 PMCID: PMC7519378 DOI: 10.1016/j.redox.2020.101723] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/31/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Increased methylglyoxal (MG) formation is associated with diabetes and its complications. In zebrafish, knockout of the main MG detoxifying system Glyoxalase 1, led to limited MG elevation but significantly elevated aldehyde dehydrogenases (ALDH) activity and aldh3a1 expression, suggesting the compensatory role of Aldh3a1 in diabetes. To evaluate the function of Aldh3a1 in glucose homeostasis and diabetes, aldh3a1−/− zebrafish mutants were generated using CRISPR-Cas9. Vasculature and pancreas morphology were analysed by zebrafish transgenic reporter lines. Corresponding reactive carbonyl species (RCS), glucose, transcriptome and metabolomics screenings were performed and ALDH activity was measured for further verification. Aldh3a1−/− zebrafish larvae displayed retinal vasodilatory alterations, impaired glucose homeostasis, which can be aggravated via pdx1 silencing induced hyperglycaemia. Unexpectedly, MG was not altered, but 4-hydroxynonenal (4-HNE), another prominent lipid peroxidation RCS exhibited high affinity with Aldh3a1, was increased in aldh3a1 mutants. 4-HNE was responsible for the retinal phenotype via pancreas disruption induced hyperglycaemia and can be rescued via l-Carnosine treatment. Furthermore, in type 2 diabetic patients, serum 4-HNE was increased and correlated with disease progression. Thus, our data suggest impaired 4-HNE detoxification and elevated 4-HNE concentration as biomarkers but also the possible inducers for diabetes, from genetic susceptibility to the pathological progression. Aldh3a1 mutant was generated using CRISPR/Cas9 and displayed impaired glucose homeostasis. Elevated 4-Hydroxynonenal (4-HNE) was responsible for hyperglycaemia in aldh3a1 mutants and was rescued by Carnosine. Patient serum 4-HNE level was correlated with HbA1c and fasting glucose. Impaired 4-HNE detoxification acts as possible inducers for diabetes, from genetic susceptibility to pathological progress.
Collapse
Affiliation(s)
- Bowen Lou
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710048, China
| | - Mike Boger
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Katrin Bennewitz
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carsten Sticht
- Center for Medical Research (ZMF), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Kopf
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Rüdiger Hell
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Zuyi Yuan
- Cardiovascular Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710048, China
| | - Peter Paul Nawroth
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz-Zentrum, München, Heidelberg, Germany
| | - Jens Kroll
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
33
|
Chen P, Zhang Y, Xu M, Chen H, Zou H, Zhang X, Tong H, You C, Wu M. Proteomic landscape of liver tissue in old male mice that are long-term treated with polysaccharides from Sargassum fusiforme. Food Funct 2020; 11:3632-3644. [PMID: 32292988 DOI: 10.1039/d0fo00187b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sargassum fusiforme is a type of brown algae and well known as a longevity promoting vegetable in Northeastern Asia. The polysaccharides derived from Sargassum fusiforme (SFPs) have been suggested as an antioxidant component for anti-aging function. However, global molecular changes in vivo by SFPs have not been fully elucidated. Here, we present a proteomics study using liver tissues of aged male mice that were fed with SFPs. Of forty-nine protein spots, thirty-eight were up-regulated and eleven were down-regulated, showing significant changes in abundance by two-dimensional gel electrophoresis. These differentially expressed proteins were mainly involved in oxidation-reduction, amino acid metabolism, and energy metabolism. Forty-six proteins were integrated into a unified network, with catalase (Cat) at the center. Intriguingly, most of the proteins were speculated as mitochondrial-located proteins. Our findings suggested that SFPs modulated antioxidant enzymes to scavenge redundant free radicals, thus preventing oxidative damage. In conclusion, our study provides a proteomic view on how SFPs have beneficial effects on the aspects of antioxidant and energy metabolism during the aging process. This study facilitates the understanding of anti-aging molecular mechanisms in polysaccharides derived from Sargassum fusiforme.
Collapse
Affiliation(s)
- Peichao Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Ya Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China. and Department of Natural Resources and Environmental Studies, University of Northern British Columbia, Prince George, BC, Canada
| | - Man Xu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Hongjun Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Huixi Zou
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Xu Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Cuiping You
- Department of Central Laboratory, Linyi People's Hospital, Shandong University, Linyi 276000, China.
| | - Mingjiang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
34
|
Voulgaridou GP, Tsochantaridis I, Tolkas C, Franco R, Giatromanolaki A, Panayiotidis MI, Pappa A. Aldehyde dehydrogenase 3A1 confers oxidative stress resistance accompanied by altered DNA damage response in human corneal epithelial cells. Free Radic Biol Med 2020; 150:66-74. [PMID: 32006654 DOI: 10.1016/j.freeradbiomed.2020.01.183] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/25/2022]
Abstract
Aldehyde dehydrogenase 3A1 is constitutively expressed in a taxon-specific manner in the cornea, where, due to its high abundance, it has been characterized as a corneal crystallin. ALDH3A1 has been proposed to be a multifaceted protein that protects cellular homeostasis through several modes of action. The present study examines the mechanisms by which ALDH3A1 exerts its cytoprotective role under conditions of oxidative stress. To this end, we have utilized an isogenic HCE-2 (human corneal epithelium) cell line pair differing in the expression of ALDH3A1. Single cell gel electrophoresis assay and H2DCFDA analysis revealed that the expression of ALDH3A1 protected HCE-2 cells from H2O2-, tert-butyl peroxide- and etoposide-induced oxidative and genotoxic effects. Furthermore, comparative qPCR analysis revealed that a panel of cell cycle (Cyclins B1, B2, D, E), apoptosis (p53, BAX, BCL-2, BCL-XL) and DNA damage response (DNA-PK, NBS1) genes were up-regulated in the ALDH3A1 expressing HCE-2 cells. Moreover, the expression profile of a variety of DNA damage signaling (DDS)-related genes, was investigated (under normal and oxidative stress conditions) by utilizing the RT2 profiler™ PCR array in both isogenic HCE-2 cell lines. Our results demonstrated that several genes associated with ATM/ATR signaling, cell cycle regulation, apoptosis and DNA damage repair were differentially expressed under all conditions tested. In conclusion, this study suggests that ALDH3A1 significantly contributes to the antioxidant defense of corneal homeostasis by maintaining DNA integrity possibly through altering the expression of specific DDS-related genes. Further studies will shed light on the precise role(s) of this multifunctional protein.
Collapse
Affiliation(s)
- Georgia-Persephoni Voulgaridou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, University Campus Dragana, 68100, Alexandroupolis, Greece
| | - Ilias Tsochantaridis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, University Campus Dragana, 68100, Alexandroupolis, Greece
| | - Christos Tolkas
- Department of Molecular Biology & Genetics, Democritus University of Thrace, University Campus Dragana, 68100, Alexandroupolis, Greece
| | - Rodrigo Franco
- Redox Biology Center, 114 VBS 0905, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Alexandra Giatromanolaki
- Department of Pathology, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Mihalis I Panayiotidis
- Department of Electron Microscopy & Molecular Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, 2371, Cyprus
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, University Campus Dragana, 68100, Alexandroupolis, Greece.
| |
Collapse
|
35
|
Subbannayya Y, Pinto SM, Mohanty V, Dagamajalu S, Prasad TSK, Murthy KR. What Makes Cornea Immunologically Unique and Privileged? Mechanistic Clues from a High-Resolution Proteomic Landscape of the Human Cornea. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:129-139. [PMID: 32125911 DOI: 10.1089/omi.2019.0190] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Success rates of corneal transplantation are particularly high owing to its unique, innate immune privilege derived from a phenomenon known as Anterior Chamber-Associated Immune Deviation (ACAID). Of note, cornea is a transparent, avascular structure that acts as a barrier along with sclera to protect the eye and contributes to optical power. Molecular and systems biology mechanisms underlying ACAID and the immunologically unique and privileged status of cornea are not well known. We report here a global unbiased proteomic profiling of the human cornea and the identification of 4824 proteins, the largest catalog of human corneal proteins identified to date. Moreover, signaling pathway analysis revealed enrichment of spliceosome, phagosome, lysosome, and focal adhesion pathways, thereby demonstrating the protective functions of corneal proteins. We observed an enrichment of neutrophil-mediated immune response processes in the cornea as well as proteins belonging to the complement and ER-Phagosome pathways that are suggestive of active immune and inflammatory surveillance response. This study provides a key expression map of the corneal proteome repertoire that should enable future translational medicine studies on the pathological conditions of the cornea and the mechanisms by which cornea immunology are governed. Molecular mechanisms of corneal immune privilege have broad relevance to understand and anticipate graft rejection in the field of organ transplantation.
Collapse
Affiliation(s)
- Yashwanth Subbannayya
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Sneha M Pinto
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Varshasnata Mohanty
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | | | - Krishna R Murthy
- Vittala International Institute of Ophthalmology, Bangalore, India.,Prabha Eye Clinic and Research Centre, Bangalore, India.,Institute of Bioinformatics, International Technology Park, Bangalore, India.,Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
36
|
Wang W, Wang C, Xu H, Gao Y. Aldehyde Dehydrogenase, Liver Disease and Cancer. Int J Biol Sci 2020; 16:921-934. [PMID: 32140062 PMCID: PMC7053332 DOI: 10.7150/ijbs.42300] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/03/2020] [Indexed: 12/19/2022] Open
Abstract
Acetaldehyde dehydrogenase 2 (ALDH2) is the key enzyme responsible for metabolism of the alcohol metabolite acetaldehyde in the liver. In addition to conversion of the acetaldehyde molecule, ALDH is also involved in other cellular functions. Recently, many studies have investigated the involvement of ALDH expression in viral hepatitis, alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), liver fibrosis, and liver cancer. Notably, ALDH2 expression has been linked with liver cancer risk, as well as pathogenesis and prognosis, and has emerged as a promising therapeutic target. Of note, approximately 8% of the world's population, and approximately 30-40% of the population in East Asia carry an inactive ALDH2 gene. This review summarizes new progress in understanding tissue-specific acetaldehyde metabolism by ALDH2 as well as the association of ALDH2 gene polymorphisms with liver disease and cancer. New research directions emerging in the field are also briefly discussed.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China
| | - Chunguang Wang
- Department of Thoracic & Cardiovascular Surgery, Second Clinical College, Jilin University, Changchun, 130041, China
| | - Hongxin Xu
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China
| | - Yanhang Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China
| |
Collapse
|
37
|
Pequerul R, Vera J, Giménez-Dejoz J, Crespo I, Coines J, Porté S, Rovira C, Parés X, Farrés J. Structural and kinetic features of aldehyde dehydrogenase 1A (ALDH1A) subfamily members, cancer stem cell markers active in retinoic acid biosynthesis. Arch Biochem Biophys 2020; 681:108256. [PMID: 31923393 DOI: 10.1016/j.abb.2020.108256] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/01/2020] [Accepted: 01/02/2020] [Indexed: 11/30/2022]
Abstract
Aldehyde dehydrogenases catalyze the NAD(P)+-dependent oxidation of aldehydes to their corresponding carboxylic acids. The three-dimensional structures of the human ALDH1A enzymes were recently obtained, while a complete kinetic characterization of them, under the same experimental conditions, is lacking. We show that the three enzymes, ALDH1A1, ALDH1A2 and ALDH1A3, have similar topologies, although with decreasing volumes in their substrate-binding pockets. The activity with aliphatic and retinoid aldehydes was characterized side-by-side, using an improved HPLC-based method for retinaldehyde. Hexanal was the most efficient substrate. ALDH1A1 displayed lower Km values with hexanal, trans-2-hexenal and citral, compared to ALDH1A2 and ALDH1A3. ALDH1A2 was the best enzyme for the lipid peroxidation product, 4-hydroxy-2-nonenal, in terms of kcat/Km. The catalytic efficiency towards all-trans and 9-cis-retinaldehyde was in general lower than for alkanals and alkenals. ALDH1A2 and ALDH1A3 showed higher catalytic efficiency for all-trans-retinaldehyde. The lower specificity of ALDH1A3 for 9-cis-retinaldehyde against the all-trans- isomer might be related to the smaller volume of its substrate-binding pocket. Magnesium inhibited ALDH1A1 and ALDH1A2, while it activated ALDH1A3, which is consistent with cofactor dissociation being the rate-limiting step for ALDH1A1 and ALDH1A2, and deacylation for ALDH1A3, with hexanal as a substrate. We mutated both ALDH1A1 (L114P) and ALDH1A2 (N475G, A476V, L477V, N478S) to mimic their counterpart substrate-binding pockets. ALDH1A1 specificity for citral was traced to residue 114 and to residues 458 to 461. Regarding retinaldehyde, the mutants did not show significant differences with their respective wild-type forms, suggesting that the mutated residues are not critical for retinoid specificity.
Collapse
Affiliation(s)
- Raquel Pequerul
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Javier Vera
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Joan Giménez-Dejoz
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Isidro Crespo
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Joan Coines
- Department of Inorganic and Organic Chemistry, Universitat de Barcelona, E-08028, Barcelona, Spain
| | - Sergio Porté
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Carme Rovira
- Department of Inorganic and Organic Chemistry, Universitat de Barcelona, E-08028, Barcelona, Spain
| | - Xavier Parés
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Jaume Farrés
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain.
| |
Collapse
|
38
|
Aryal S, Anand D, Hernandez FG, Weatherbee BAT, Huang H, Reddy AP, Wilmarth PA, David LL, Lachke SA. MS/MS in silico subtraction-based proteomic profiling as an approach to facilitate disease gene discovery: application to lens development and cataract. Hum Genet 2019; 139:151-184. [PMID: 31797049 DOI: 10.1007/s00439-019-02095-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/24/2019] [Indexed: 12/20/2022]
Abstract
While the bioinformatics resource-tool iSyTE (integrated Systems Tool for Eye gene discovery) effectively identifies human cataract-associated genes, it is currently based on just transcriptome data, and thus, it is necessary to include protein-level information to gain greater confidence in gene prioritization. Here, we expand iSyTE through development of a novel proteome-based resource on the lens and demonstrate its utility in cataract gene discovery. We applied high-throughput tandem mass spectrometry (MS/MS) to generate a global protein expression profile of mouse lens at embryonic day (E)14.5, which identified 2371 lens-expressed proteins. A major challenge of high-throughput expression profiling is identification of high-priority candidates among the thousands of expressed proteins. To address this problem, we generated new MS/MS proteome data on mouse whole embryonic body (WB). WB proteome was then used as a reference dataset for performing "in silico WB-subtraction" comparative analysis with the lens proteome, which effectively identified 422 proteins with lens-enriched expression at ≥ 2.5 average spectral counts, ≥ 2.0 fold enrichment (FDR < 0.01) cut-off. These top 20% candidates represent a rich pool of high-priority proteins in the lens including known human cataract-linked genes and many new potential regulators of lens development and homeostasis. This rich information is made publicly accessible through iSyTE (https://research.bioinformatics.udel.edu/iSyTE/), which enables user-friendly visualization of promising candidates, thus making iSyTE a comprehensive tool for cataract gene discovery.
Collapse
Affiliation(s)
- Sandeep Aryal
- Department of Biological Sciences, University of Delaware, 105 The Green, Delaware Avenue, 236 Wolf Hall, Newark, DE, USA
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, 105 The Green, Delaware Avenue, 236 Wolf Hall, Newark, DE, USA
| | - Francisco G Hernandez
- Department of Biological Sciences, University of Delaware, 105 The Green, Delaware Avenue, 236 Wolf Hall, Newark, DE, USA
| | - Bailey A T Weatherbee
- Department of Biological Sciences, University of Delaware, 105 The Green, Delaware Avenue, 236 Wolf Hall, Newark, DE, USA
| | - Hongzhan Huang
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, 19716, USA
| | - Ashok P Reddy
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Larry L David
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Salil A Lachke
- Department of Biological Sciences, University of Delaware, 105 The Green, Delaware Avenue, 236 Wolf Hall, Newark, DE, USA.
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
39
|
Rathinam S, Daniel I, Kuppamuthu D, Jayapal JM. Molecular Mimicry between Betaine Aldehyde Dehydrogenase of Leptospira and Retinal Dehydrogenase 1 of Human Lens: A Potential Trigger for Cataract Formation in Leptospiral Uveitis Patients. Ocul Immunol Inflamm 2019; 29:579-586. [PMID: 31746662 DOI: 10.1080/09273948.2019.1687732] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Purpose: Rapidly progressing cataract is one of the ocular manifestations in leptospiral uveitis patients. We examined whether molecular mimicry between the leptospira antigens and lens proteins exists that could result in cataract in these patients.Methods: Immunoblot analysis using patient sera was done with proteins from normal lens and cataract lens from leptospiral uveitis patients and the cross-reacting lens proteins were identified by mass spectrometry analysis.Results: Retinal dehydrogenase 1 and crystallins (α-B, α-A2, β-B2), were recognized by the antibodies in the serum of leptospiral uveitis patients. And, retinal dehydrogenase 1 is homologous to the leptospiral protein, betaine aldehyde dehydrogenase.Conclusions: Leptospiral uveitis patient serum contains antibodies that cross-react with multiple lens proteins that have a role in maintaining lens transparency. And, these antibodies could act as a potential trigger for cataractogenesis.
Collapse
Affiliation(s)
| | - Irene Daniel
- Proteomics Department, Aravind Medical Research Foundation, Madurai, India
| | | | | |
Collapse
|
40
|
Calleja LF, Belmont-Díaz JA, Medina-Contreras O, Quezada H, Yoval-Sánchez B, Campos-García J, Rodríguez-Zavala JS. Omeprazole as a potent activator of human cytosolic aldehyde dehydrogenase ALDH1A1. Biochim Biophys Acta Gen Subj 2019; 1864:129451. [PMID: 31678145 DOI: 10.1016/j.bbagen.2019.129451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/30/2019] [Accepted: 10/23/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Accumulation of lipid aldehydes plays a key role in the etiology of human diseases where high levels of oxidative stress are generated. In this regard, activation of aldehyde dehydrogenases (ALDHs) prevents oxidative tissue damage during ischemia-reperfusion processes. Although omeprazole is used to reduce stomach gastric acid production, in the present work this drug is described as the most potent activator of human ALDH1A1 reported yet. METHODS Docking analysis was performed to predict the interactions of omeprazole with the enzyme. Recombinant human ALDH1A1 was used to assess the effect of omeprazole on the kinetic properties. Temperature treatment and mass spectrometry were conducted to address the nature of binding of the activator to the enzyme. Finally, the effect of omeprazole was evaluated in an in vivo model of oxidative stress, using E. coli cells expressing the human ALDH1A1. RESULTS Omeprazole interacted with the aldehyde binding site, increasing 4-6 fold the activity of human ALDH1A1, modified the kinetic properties, altering the order of binding of substrates and release of products, and protected the enzyme from inactivation by lipid aldehydes. Furthermore, omeprazole protected E. coli cells over-expressing ALDH1A1 from the effects of oxidative stress generated by H2O2 exposure, reducing the levels of lipid aldehydes and preserving ALDH activity. CONCLUSION Omeprazole can be repositioned as a potent activator of human ALDH1A1 and may be proposed for its use in therapeutic strategies, to attenuate the damage generated during oxidative stress events occurring in different human pathologies.
Collapse
Affiliation(s)
- Luis Francisco Calleja
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico 14080
| | | | - Oscar Medina-Contreras
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City, Mexico 06720
| | - Héctor Quezada
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City, Mexico 06720
| | - Belem Yoval-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico 14080
| | - Jesús Campos-García
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mich., Mexico 58030
| | | |
Collapse
|
41
|
Chacón‐Solano E, León C, Díaz F, García‐García F, García M, Escámez M, Guerrero‐Aspizua S, Conti C, Mencía Á, Martínez‐Santamaría L, Llames S, Pévida M, Carbonell‐Caballero J, Puig‐Butillé J, Maseda R, Puig S, de Lucas R, Baselga E, Larcher F, Dopazo J, del Río M. Fibroblast activation and abnormal extracellular matrix remodelling as common hallmarks in three cancer-prone genodermatoses. Br J Dermatol 2019; 181:512-522. [PMID: 30693469 PMCID: PMC6850467 DOI: 10.1111/bjd.17698] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Recessive dystrophic epidermolysis bullosa (RDEB), Kindler syndrome (KS) and xeroderma pigmentosum complementation group C (XPC) are three cancer-prone genodermatoses whose causal genetic mutations cannot fully explain, on their own, the array of associated phenotypic manifestations. Recent evidence highlights the role of the stromal microenvironment in the pathology of these disorders. OBJECTIVES To investigate, by means of comparative gene expression analysis, the role played by dermal fibroblasts in the pathogenesis of RDEB, KS and XPC. METHODS We conducted RNA-Seq analysis, which included a thorough examination of the differentially expressed genes, a functional enrichment analysis and a description of affected signalling circuits. Transcriptomic data were validated at the protein level in cell cultures, serum samples and skin biopsies. RESULTS Interdisease comparisons against control fibroblasts revealed a unifying signature of 186 differentially expressed genes and four signalling pathways in the three genodermatoses. Remarkably, some of the uncovered expression changes suggest a synthetic fibroblast phenotype characterized by the aberrant expression of extracellular matrix (ECM) proteins. Western blot and immunofluorescence in situ analyses validated the RNA-Seq data. In addition, enzyme-linked immunosorbent assay revealed increased circulating levels of periostin in patients with RDEB. CONCLUSIONS Our results suggest that the different causal genetic defects converge into common changes in gene expression, possibly due to injury-sensitive events. These, in turn, trigger a cascade of reactions involving abnormal ECM deposition and underexpression of antioxidant enzymes. The elucidated expression signature provides new potential biomarkers and common therapeutic targets in RDEB, XPC and KS. What's already known about this topic? Recessive dystrophic epidermolysis bullosa (RDEB), Kindler syndrome (KS) and xeroderma pigmentosum complementation group C (XPC) are three genodermatoses with high predisposition to cancer development. Although their causal genetic mutations mainly affect epithelia, the dermal microenvironment likely contributes to the physiopathology of these disorders. What does this study add? We disclose a large overlapping transcription profile between XPC, KS and RDEB fibroblasts that points towards an activated phenotype with high matrix-synthetic capacity. This common signature seems to be independent of the primary causal deficiency, but reflects an underlying derangement of the extracellular matrix via transforming growth factor-β signalling activation and oxidative state imbalance. What is the translational message? This study broadens the current knowledge about the pathology of these diseases and highlights new targets and biomarkers for effective therapeutic intervention. It is suggested that high levels of circulating periostin could represent a potential biomarker in RDEB.
Collapse
|
42
|
Nedelec B, Rozet JM, Fares Taie L. Genetic architecture of retinoic-acid signaling-associated ocular developmental defects. Hum Genet 2019; 138:937-955. [DOI: 10.1007/s00439-019-02052-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022]
|
43
|
McKinney A, Lindberg OR, Engler JR, Chen KY, Kumar A, Gong H, Lu KV, Simonds EF, Cloughesy TF, Liau LM, Prados M, Bollen AW, Berger MS, Shieh JTC, James CD, Nicolaides TP, Yong WH, Lai A, Hegi ME, Weiss WA, Phillips JJ. Mechanisms of Resistance to EGFR Inhibition Reveal Metabolic Vulnerabilities in Human GBM. Mol Cancer Ther 2019; 18:1565-1576. [PMID: 31270152 DOI: 10.1158/1535-7163.mct-18-1330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 05/10/2019] [Accepted: 06/28/2019] [Indexed: 12/23/2022]
Abstract
Amplification of the epidermal growth factor receptor gene (EGFR) represents one of the most commonly observed genetic lesions in glioblastoma (GBM); however, therapies targeting this signaling pathway have failed clinically. Here, using human tumors, primary patient-derived xenografts (PDX), and a murine model for GBM, we demonstrate that EGFR inhibition leads to increased invasion of tumor cells. Further, EGFR inhibitor-treated GBM demonstrates altered oxidative stress, with increased lipid peroxidation, and generation of toxic lipid peroxidation products. A tumor cell subpopulation with elevated aldehyde dehydrogenase (ALDH) levels was determined to comprise a significant proportion of the invasive cells observed in EGFR inhibitor-treated GBM. Our analysis of the ALDH1A1 protein in newly diagnosed GBM revealed detectable ALDH1A1 expression in 69% (35/51) of the cases, but in relatively low percentages of tumor cells. Analysis of paired human GBM before and after EGFR inhibitor therapy showed an increase in ALDH1A1 expression in EGFR-amplified tumors (P < 0.05, n = 13 tumor pairs), and in murine GBM ALDH1A1-high clones were more resistant to EGFR inhibition than ALDH1A1-low clones. Our data identify ALDH levels as a biomarker of GBM cells with high invasive potential, altered oxidative stress, and resistance to EGFR inhibition, and reveal a therapeutic target whose inhibition should limit GBM invasion.
Collapse
Affiliation(s)
- Andrew McKinney
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Olle R Lindberg
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Jane R Engler
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Katharine Y Chen
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Anupam Kumar
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Henry Gong
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Kan V Lu
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Erin F Simonds
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California.,Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Linda M Liau
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Michael Prados
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Andrew W Bollen
- Department of Pathology, Division of Neuropathology, University of California, San Francisco, San Francisco, California
| | - Mitchel S Berger
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Joseph T C Shieh
- Division of Medical Genetics, Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - C David James
- Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Theodore P Nicolaides
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California.,Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California
| | - William H Yong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Albert Lai
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Monika E Hegi
- Neuroscience Research Center and Service of Neurosurgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - William A Weiss
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California.,Department of Neurology, University of California, San Francisco, San Francisco, California.,Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California. .,Department of Pathology, Division of Neuropathology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
44
|
Marshall S, Chen Y, Singh S, Berrios-Carcamo P, Heit C, Apostolopoulos N, Golla JP, Thompson DC, Vasiliou V. Engineered Animal Models Designed for Investigating Ethanol Metabolism, Toxicity and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1032:203-221. [PMID: 30362100 PMCID: PMC6743736 DOI: 10.1007/978-3-319-98788-0_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Excessive consumption of alcohol is a leading cause of lifestyle-induced morbidity and mortality worldwide. Although long-term alcohol abuse has been shown to be detrimental to the liver, brain and many other organs, our understanding of the exact molecular mechanisms by which this occurs is still limited. In tissues, ethanol is metabolized to acetaldehyde (mainly by alcohol dehydrogenase and cytochrome p450 2E1) and subsequently to acetic acid by aldehyde dehydrogenases. Intracellular generation of free radicals and depletion of the antioxidant glutathione (GSH) are believed to be key steps involved in the cellular pathogenic events caused by ethanol. With continued excessive alcohol consumption, further tissue damage can result from the production of cellular protein and DNA adducts caused by accumulating ethanol-derived aldehydes. Much of our understanding about the pathophysiological consequences of ethanol metabolism comes from genetically-engineered mouse models of ethanol-induced tissue injury. In this review, we provide an update on the current understanding of important mouse models in which ethanol-metabolizing and GSH-synthesizing enzymes have been manipulated to investigate alcohol-induced disease.
Collapse
Affiliation(s)
- Stephanie Marshall
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Surendra Singh
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Pablo Berrios-Carcamo
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
- Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claire Heit
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, CO, USA
| | - Nicholas Apostolopoulos
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Jaya Prakash Golla
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - David C Thompson
- Department of Clinical Pharmacy, Skaggs School of Pharmacy, University of Colorado, Aurora, CO, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA.
| |
Collapse
|
45
|
Srividya G, Angayarkanni N, Iyer G, Srinivasan B, Agarwal S. Altered retinoid metabolism gene expression in chronic Stevens-Johnson syndrome. Br J Ophthalmol 2019; 103:1015-1023. [DOI: 10.1136/bjophthalmol-2018-312849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 02/09/2019] [Accepted: 03/09/2019] [Indexed: 12/19/2022]
Abstract
BackgroundStevens-Johnson syndrome (SJS), a blistering disorder of the skin and mucous membrane, leads to ocular morbidity in >60% of cases. Retinoids are vital micronutrients for vision, regulating corneal and conjunctival cell proliferation, differentiation and immune function. This prospective case–control study probed for alterations in retinoid metabolism by evaluating retinoic acid receptor signalling in the conjunctival cells of patients with SJS.MethodsImprints were collected from the bulbar conjunctiva of patients with chronic SJS. The gene expression of retinoic acid receptors, namely, RXRA, RARA, RARG, RORA; the fibrosis marker TGFβ and its receptor TGFβRII; the transcription factors PPAR-γ, STRA6 and Stat3; the enzymes aldehyde dehydrogenase (ALDH1a1), alpha-1 antitrypsin (A1AT); and the Cyp genes Cyp26a1 and Cyp26b1 were assessed by quantitative PCR in patients with SJS pre-mucous (n = 34) and post-mucous membrane graft (MMG) intervention (n=19) in comparison with age-matched/sex-matched healthy controls (n=20). Western blot analysis of ALDH1a1, RARA and RARG were done in the conjunctival imprint cells.ResultsThe transcript levels of ALDH1a1, RXRA, RORA, STRA6, Cyp26a1 and Cyp26b1 were decreased around 4, 26, 17, 129, 9 and 8 folds, respectively, and RARA, RARG, PPAR-γ, TGFβ, TGFβRII were increased by 12, 15, 51, 16 and 87 folds, respectively, in SJS conjunctiva at the pre-MMG stage. The changes in RORA, Cyp26a1, Cyp26b1, RARA and Stat3 were statistically significant (p<0.05). Changes in protein expression of ALDH1a1, RARA and RARG supported the gene expression changes.ConclusionsThe study provides the first experimental insight into the role of retinoid metabolism in the ocular sequelae of chronic SJS.
Collapse
|
46
|
Wang Y, Wang CH, Zhang YF, Zhu L, Lei HM, Tang YB. UPLC-MS-based metabolomics reveals metabolic dysregulation in ALDH1A1-overexpressed lung adenocarcinoma cells. Metabolomics 2019; 15:52. [PMID: 30911937 DOI: 10.1007/s11306-019-1514-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/18/2019] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Specific oncogenotypes can produce distinct metabolic changes in cancer. Recently it is considered that metabolic reprograming contributes heavily to drug resistance. Aldehyde dehydrogenase 1A1 (ALDH1A1), is overexpressed in drug resistant lung adenocarcinomas and may be the cause of acquired drug resistance. However, how ALDH1A1 affects metabolic profiling in lung adenocarcinoma cells remains elusive. OBJECTIVE We sought to investigate metabolic alterations induced by ALDH1A1 in lung adenocarcinoma in order to better understand the reprogramming and metabolic mechanism of resistance induced by ALDH1A1. METHODS Metabolic alterations in lung adenocarcinoma HCC827-ALDH1A1 cells were analyzed by ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS). HCC827-ALDH1A1 metabolic signatures were extracted by univariate and multivariate statistical analysis. Furthermore, metabolite enrichment analysis and pathway analysis were performed using MetaboAnalyst 4.0 software. RESULTS Twenty-two metabolites were positively identified using authentic standards, including uridine monophosphate (UMP), uridine diphosphate (UDP), adenosine diphosphate (ADP), malic acid, malonyl-coenzyme A, nicotinamide adenine dinucleotide (NAD), coenzyme A and so on. Furthermore, metabolic pathway analysis revealed several dysregulated pathways in HCC827-ALDH1A1 cells, including nucleotide metabolism, urea cycle, tricarboxylic acid (TCA) cycle, and glycerol phospholipid metabolism etc. CONCLUSION: Lung cancer is the most frequent cause of cancer-related deaths worldwide. Nearly all patients eventually undergo disease progression due to acquired resistance. Mechanisms of biological acquired resistance need to be identified. Our study identified altered metabolites in HCC827-ALDH1A1 cells, enhancing our knowledge of lung adenocarcinoma metabolic alterations induced by ALDH1A1, creating a novel therapeutic pathway. These metabolic signatures of ALDH1A1 overexpression may shed light on molecular mechanisms in drug-resistant tumors, and on candidate drug targets. Furthermore, new molecular targets may provide the foundation for potential anticancer strategies for lung cancer therapy.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Cong-Hui Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu-Fei Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hui-Min Lei
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ya-Bin Tang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
47
|
Rodríguez-Zavala JS, Calleja LF, Moreno-Sánchez R, Yoval-Sánchez B. Role of Aldehyde Dehydrogenases in Physiopathological Processes. Chem Res Toxicol 2019; 32:405-420. [PMID: 30628442 DOI: 10.1021/acs.chemrestox.8b00256] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Many different diseases are associated with oxidative stress. One of the main consequences of oxidative stress at the cellular level is lipid peroxidation, from which toxic aldehydes may be generated. Below their toxicity thresholds, some aldehydes are involved in signaling processes, while others are intermediaries in the metabolism of lipids, amino acids, neurotransmitters, and carbohydrates. Some aldehydes ubiquitously distributed in the environment, such as acrolein or formaldehyde, are extremely toxic to the cell. On the other hand, aldehyde dehydrogenases (ALDHs) are able to detoxify a wide variety of aldehydes to their corresponding carboxylic acids, thus helping to protect from oxidative stress. ALDHs are located in different subcellular compartments such as cytosol, mitochondria, nucleus, and endoplasmic reticulum. The aim of this review is to analyze, and highlight, the role of different ALDH isoforms in the detoxification of aldehydes generated in processes that involve high levels of oxidative stress. The ALDH physiological relevance becomes evident by the observation that their expression and activity are enhanced in different pathologies that involve oxidative stress such as neurodegenerative disorders, cardiopathies, atherosclerosis, and cancer as well as inflammatory processes. Furthermore, ALDH mutations bring about several disorders in the cell. Thus, understanding the mechanisms by which these enzymes participate in diverse cellular processes may lead to better contend with the damage caused by toxic aldehydes in different pathologies by designing modulators and/or protocols to modify their activity or expression.
Collapse
Affiliation(s)
| | | | - Rafael Moreno-Sánchez
- Departamento de Bioquímica , Instituto Nacional de Cardiología , México 14080 , México
| | - Belem Yoval-Sánchez
- Departamento de Bioquímica , Instituto Nacional de Cardiología , México 14080 , México
| |
Collapse
|
48
|
Toledo-Guzmán ME, Hernández MI, Gómez-Gallegos ÁA, Ortiz-Sánchez E. ALDH as a Stem Cell Marker in Solid Tumors. Curr Stem Cell Res Ther 2019; 14:375-388. [PMID: 30095061 DOI: 10.2174/1574888x13666180810120012] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023]
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme that participates in important cellular mechanisms as aldehyde detoxification and retinoic acid synthesis; moreover, ALDH activity is involved in drug resistance, a characteristic of cancer stem cells (CSCs). Even though ALDH is found in stem cells, CSCs and progenitor cells, this enzyme has been successfully used to identify and isolate cell populations with CSC properties from several tumor origins. ALDH is allegedly involved in cell differentiation through its product, retinoic acid. However, direct or indirect ALDH inhibition, using specific inhibitors or retinoic acid, has shown a reduction in ALDH activity, along with the loss of stem cell traits, reduction of cell proliferation, invasion, and drug sensitization. For these reasons, ALDH and retinoic acid are promising therapeutic targets. This review summarizes the current evidence for ALDH as a CSCs marker in solid tumors, as well as current knowledge about the functional roles of ALDH in CSCs. We discuss the controversy of ALDH activity to maintain CSC stemness, or conversely, to promote cell differentiation. Finally, we review the advances in using ALDH inhibitors as anti-cancer drugs.
Collapse
Affiliation(s)
- Mariel E Toledo-Guzmán
- Departamento de Bioquimica, Laboratorio de Terapia Genica, Escuela Nacional de Ciencias Biologicas, Posgrado de Biomedicina y Biotecnologia Molecular, Instituto Politecnico Nacional, Mexico City, Mexico
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
| | - Miguel Ibañez Hernández
- Departamento de Bioquimica, Laboratorio de Terapia Genica, Escuela Nacional de Ciencias Biologicas, Posgrado de Biomedicina y Biotecnologia Molecular, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Ángel A Gómez-Gallegos
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
- Posgrado de Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Av San Fernando 22, Colonia Seccion XVI, Tlalpan 14080, Mexico City, Mexico
| |
Collapse
|
49
|
Luo YX, Wang XY, Huang YJ, Fang SH, Wu J, Zhang YB, Xiong TQ, Yang C, Shen JG, Sang CL, Wang Q, Fang JS. Systems pharmacology-based investigation of Sanwei Ganjiang Prescription: related mechanisms in liver injury. Chin J Nat Med 2018; 16:756-765. [PMID: 30322609 DOI: 10.1016/s1875-5364(18)30115-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Liver injury remains a significant global health problem and has a variety of causes, including oxidative stress (OS), inflammation, and apoptosis of liver cells. There is currently no curative therapy for this disorder. Sanwei Ganjiang Prescription (SWGJP), derived from traditional Chinese medicine (TCM), has shown its effectiveness in long-term liver damage therapy, although the underlying molecular mechanisms are still not fully understood. To explore the underlining mechanisms of action for SWGJP in liver injury from a holistic view, in the present study, a systems pharmacology approach was developed, which involved drug target identification and multilevel data integration analysis. Using a comprehensive systems approach, we identified 43 candidate compounds in SWGJP and 408 corresponding potential targets. We further deciphered the mechanisms of SWGJP in treating liver injury, including compound-target network analysis, target-function network analysis, and integrated pathways analysis. We deduced that SWGJP may protect hepatocytes through several functional modules involved in liver injury integrated-pathway, such as Nrf2-dependent anti-oxidative stress module. Notably, systems pharmacology provides an alternative way to investigate the complex action mode of TCM.
Collapse
Affiliation(s)
- Yun-Xia Luo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xin-Yue Wang
- Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yu-Jie Huang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shu-Huan Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jun Wu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yong-Bin Zhang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Tian-Qin Xiong
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Cong Yang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jian-Gang Shen
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Chuan-Lan Sang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Jian-Song Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
50
|
Cvekl A, Zhao Y, McGreal R, Xie Q, Gu X, Zheng D. Evolutionary Origins of Pax6 Control of Crystallin Genes. Genome Biol Evol 2018; 9:2075-2092. [PMID: 28903537 PMCID: PMC5737492 DOI: 10.1093/gbe/evx153] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2017] [Indexed: 12/19/2022] Open
Abstract
The birth of novel genes, including their cell-specific transcriptional control, is a major source of evolutionary innovation. The lens-preferred proteins, crystallins (vertebrates: α- and β/γ-crystallins), provide a gateway to study eye evolution. Diversity of crystallins was thought to originate from convergent evolution through multiple, independent formation of Pax6/PaxB-binding sites within the promoters of genes able to act as crystallins. Here, we propose that αB-crystallin arose from a duplication of small heat shock protein (Hspb1-like) gene accompanied by Pax6-site and heat shock element (HSE) formation, followed by another duplication to generate the αA-crystallin gene in which HSE was converted into another Pax6-binding site. The founding β/γ-crystallin gene arose from the ancestral Hspb1-like gene promoter inserted into a Ca2+-binding protein coding region, early in the cephalochordate/tunicate lineage. Likewise, an ancestral aldehyde dehydrogenase (Aldh) gene, through multiple gene duplications, expanded into a multigene family, with specific genes expressed in invertebrate lenses (Ω-crystallin/Aldh1a9) and both vertebrate lenses (η-crystallin/Aldh1a7 and Aldh3a1) and corneas (Aldh3a1). Collectively, the present data reconstruct the evolution of diverse crystallin gene families.
Collapse
Affiliation(s)
- Ales Cvekl
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York.,Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Yilin Zhao
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Rebecca McGreal
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York.,Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Qing Xie
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York.,Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Xun Gu
- Program in Bioinformatics and Computational Biology, Department of Genetics, Development, and Cell Biology, Iowa State University
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York.,Department of Neurology, Albert Einstein College of Medicine, Bronx, New York.,Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|