1
|
Wang YN, Liu S. The role of ALDHs in lipid peroxidation-related diseases. Int J Biol Macromol 2024; 288:138760. [PMID: 39674477 DOI: 10.1016/j.ijbiomac.2024.138760] [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: 04/15/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Lipid peroxidation presents the oxidative degradation of polyunsaturated fatty acids lincited by reactive species. Excessive accumulation of lipid peroxidation byproducts, including 4-hydroxy-2-nonenal (4-HNE) and malondialdehyde (MDA), causes protein dysfunction and various illnesses. Aldehyde dehydrogenases (ALDHs) catalyze the metabolism of both endogenous and exogenous aldehydes. These enzymes participate in detoxification and intermediary metabolism. Contemporary research has affirmed the involvement of both enzymatic and non-enzymatic pathways of ALDHs in modulating the evolution of diseases associated with lipid peroxidation. This review provides an overview of the biological functions and clinical implications concerning the enzymatic and non-enzymatic pathways of ALDHs in diseases related to lipid peroxidation, such as, non-alcoholic fatty liver disease (NAFLD), atherosclerosis, and type 2 diabetes (T2DM). Furthermore, the activators or inhibitors of ALDHs represent a promising therapeutic strategy for lipid peroxidation-related diseases.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of Implantology & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China; Suzhou Research Institute, Shandong University, Suzhou, Jiangsu 215123, China
| | - Shiyue Liu
- Department of Implantology & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China.
| |
Collapse
|
2
|
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
|
3
|
Pan J, Dong Y, Zou Z, Gu T, Chen L, Li K, Wang L, Shi Q. Serum proteome profiling of plateau acclimatization in men using Olink proteomics approach. Physiol Rep 2024; 12:e70091. [PMID: 39725655 DOI: 10.14814/phy2.70091] [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: 05/02/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 12/28/2024] Open
Abstract
Plateau acclimatization involves adaptive changes in the body's neurohumoral regulation and metabolic processes due to hypoxic conditions at high altitudes. This study utilizes Olink targeted proteomics to analyze serum protein expression differences in Han Chinese individuals acclimatized for 6 months-1 year at 4500 and 5300 m altitudes, compared to those residing at sea level. The objective is to elucidate the proteins' roles in tissue and cellular adaptation to hypoxia. We identified 54 metabolism-related differentially expressed proteins (DEPs) in the serum of the high-altitude group versus the sea-level group, comprising 20 significantly upregulated and 34 downregulated proteins. Notably, 2 proteins were upregulated and 11 downregulated at both 4500 and 5300 m altitudes. The top three protein correlations among DEPs included CRKL with CA13, RNASE3 with NADK, and NADK with APEX1, alongside APLP1 with CTSH, CTSH with SOST, and CTSH with NT-proBNP in inverse correlations. KEGG enrichment analysis indicated significant DEP involvement in various metabolic pathways, particularly those associated with hypoxic cellular metabolism like glycolysis/gluconeogenesis and the HIF-1 signaling pathway. Correlation with clinical phenotypes showed positive associations of SOST, RNASE3, CA13, NADK, and CRKL with SaO2 and negative correlations with Hemoglobin and Hematocrit; ALDH1A1 positively correlated with Triglyceride; and SDC4 inversely correlated with Uric acid levels. This study provides insights into specific DEPs linked to metabolic adaptations in high-altitude acclimatized individuals, offering a foundation for understanding acclimatization mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Jingyu Pan
- Medical College of Shihezi University, Urumqi, Xinjiang, China
- Clinical Laboratory Diagnostic Center, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
| | - Yue Dong
- The Second Department of Cadre Health Care, Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhihao Zou
- Department of Neurosurgery, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
| | - Tianyan Gu
- Clinical Laboratory Diagnostic Center, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
- Graduate School of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ling Chen
- Clinical Laboratory Diagnostic Center, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
| | - Kai Li
- Clinical Laboratory Diagnostic Center, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
| | - Li Wang
- Clinical Laboratory Diagnostic Center, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
| | - Qinghai Shi
- Clinical Laboratory Diagnostic Center, General Hospital of Xinjiang Military Command, Urumqi, Xinjiang, China
| |
Collapse
|
4
|
Artasensi A, Mazzotta S, Sanz I, Lin L, Vistoli G, Fumagalli L, Regazzoni L. Exploring Secondary Amine Carnosine Derivatives: Design, Synthesis, and Properties. Molecules 2024; 29:5083. [PMID: 39519724 PMCID: PMC11547551 DOI: 10.3390/molecules29215083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Carnosine is a naturally occurring dipeptide that has been advocated by some authors as an interesting scaffold for the development of potential therapeutic agents in view of the positive outcomes of its supplementation in animal models of human diseases. Its mode of action seems to depend on the quenching of toxic electrophiles, such as 4-hydroxynonenal (HNE). However, carnosine's bioavailability in humans is lower than that in other mammals. The main reason for such an unfavorable pharmacokinetic profile is the activity of the enzyme human serum carnosinase (E.C. 3.4.13.20), which rapidly hydrolyzes carnosine upon absorption. Therefore, some studies have focused on the design of carnosinase-resistant derivatives that retain binding activity toward toxic electrophiles. Nevertheless, the structural modification of the N-terminus amino group of carnosine has rarely been considered, possibly because of its key role in the electrophile scavenging mechanism. This was proven, since some carnosine N-terminus modification generated inactive compounds, despite some derivatives retaining oral bioavailability and gaining resistance to carnosinase hydrolysis. Herein, we therefore report a study aimed at exploring whether the amino group of carnosine can be conveniently modified to develop carnosinase-resistant derivatives retaining the dipeptide activity toward toxic electrophiles.
Collapse
Affiliation(s)
- Angelica Artasensi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Sarah Mazzotta
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133 Milan, Italy
| | - Ines Sanz
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Licheng Lin
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Giulio Vistoli
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Laura Fumagalli
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| | - Luca Regazzoni
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via L. Mangiagalli 25, 20133 Milan, Italy
| |
Collapse
|
5
|
Monroe TB, Hertzel AV, Dickey DM, Hagen T, Santibanez SV, Berdaweel IA, Halley C, Puchalska P, Anderson EJ, Camell CD, Robbins PD, Bernlohr DA. Lipid peroxidation products induce carbonyl stress, mitochondrial dysfunction, and cellular senescence in human and murine cells. Aging Cell 2024:e14367. [PMID: 39394673 DOI: 10.1111/acel.14367] [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: 03/24/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 10/14/2024] Open
Abstract
Lipid enals are electrophilic products of lipid peroxidation that induce genotoxic and proteotoxic stress by covalent modification of DNA and proteins, respectively. As lipid enals accumulate to substantial amounts in visceral adipose during obesity and aging, we hypothesized that biogenic lipid enals may represent an endogenously generated, and therefore physiologically relevant, senescence inducers. To that end, we identified that 4-hydroxynonenal (4-HNE), 4-hydroxyhexenal (4-HHE) or 4-oxo-2-nonenal (4-ONE) initiate the cellular senescence program of IMR90 fibroblasts and murine adipose stem cells. In such cells, lipid enals induced accumulation of γH2AX foci, increased p53 signaling, enhanced expression of p21Cip1, and upregulated the expression and secretion of numerous cytokines, chemokines, and regulatory factors independently from NF-κB activation. Concomitantly, lipid enal treatment resulted in covalent modification of mitochondrial proteins, reduced mitochondrial spare respiratory capacity, altered nucleotide pools, and increased the phosphorylation of AMP kinase. Lipid-induced senescent cells upregulated BCL2L1 (Bcl-xL) and BCL2L2 (Bcl-w). and were resistant to apoptosis while pharmacologic inhibition of BAX/BAK macropores attenuated lipid-induced senescence. In situ, the 4-HNE scavenger L-carnosine ameliorated the development of the cellular senescence, while in visceral fat of obese C57BL/6J mice, L-carnosine reduced the abundance of 4-HNE-modified proteins and blunted the expression of senescence biomarkers CDKN1A (p21Cip1), PLAUR, BCL2L1, and BCL2L2. Taken together, the results suggest that lipid enals are endogenous regulators of cellular senescence and that biogenic lipid-induced senescence (BLIS) may represent a mechanistic link between oxidative stress and age-dependent pathologies.
Collapse
Affiliation(s)
- T Blake Monroe
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Deborah M Dickey
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Thomas Hagen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Simon Vergara Santibanez
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Islam A Berdaweel
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA
| | - Catherine Halley
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Patrycja Puchalska
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Ethan J Anderson
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA
| | - Christina D Camell
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
- Institute for the Biology of Aging and Metabolism, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
- Institute for the Biology of Aging and Metabolism, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota, USA
| |
Collapse
|
6
|
Li SY, Tsai MT, Kuo YM, Yang HM, Tong ZJ, Cheng HW, Lin CC, Wang HT. Aldehyde dehydrogenase 2 preserves kidney function by countering acrolein-induced metabolic and mitochondrial dysfunction. JCI Insight 2024; 9:e179871. [PMID: 39226171 PMCID: PMC11466184 DOI: 10.1172/jci.insight.179871] [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: 02/26/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024] Open
Abstract
The prevalence of chronic kidney disease (CKD) varies by race because of genetic and environmental factors. The Glu504Lys polymorphism in aldehyde dehydrogenase 2 (ALDH2), commonly observed among East Asian people, alters the enzyme's function in detoxifying alcohol-derived aldehydes, affecting kidney function. This study investigated the association between variations in ALDH2 levels within the kidney and the progression of kidney fibrosis. Our clinical data indicate that diminished ALDH2 levels are linked to worse CKD outcomes, with correlations between ALDH2 expression, estimated glomerular filtration rate, urinary levels of acrolein - an aldehyde metabolized by ALDH2 - and fibrosis severity. In mouse models of unilateral ureteral obstruction and folic acid nephropathy, reduced ALDH2 levels and elevated acrolein were observed in kidneys, especially in ALDH2 Glu504Lys-knockin mice. Mechanistically, acrolein modifies pyruvate kinase M2, leading to its nuclear translocation and coactivation of HIF-1α, shifting cellular metabolism to glycolysis, disrupting mitochondrial function, and contributing to tubular damage and the progression of kidney fibrosis. Enhancing ALDH2 expression through adeno-associated virus vectors reduced acrolein and mitigated fibrosis in both WT and Glu504Lys-knockin mice. These findings underscore the potential therapeutic significance of targeting the dynamic interaction between ALDH2 and acrolein in CKD.
Collapse
Affiliation(s)
- Szu-Yuan Li
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine and
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine and
| | - Yu-Ming Kuo
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hui-Min Yang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Zhen-Jie Tong
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Wei Cheng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine and
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctor Degree Program in Toxicology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
7
|
Venton G, Colle J, Tichadou A, Quessada J, Baier C, Labiad Y, Perez M, De Lassus L, Loosveld M, Arnoux I, Abbou N, Ceylan I, Martin G, Costello R. Reactive oxygen species and aldehyde dehydrogenase 1A as prognosis and theragnostic biomarker in acute myeloid leukaemia patients. J Cell Mol Med 2024; 28:e70011. [PMID: 39392121 PMCID: PMC11467733 DOI: 10.1111/jcmm.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/15/2024] [Accepted: 08/06/2024] [Indexed: 10/12/2024] Open
Abstract
Acute myeloid leukaemia (AML) remains a major unmet medical, despite recent progress in targeted molecular therapies. One aspect of leukaemic cell resistance to chemotherapy is the development of clones with increased capacity to respond to cellular stress and the production of reactive oxygen species (ROS), thanks in particular to a high aldehyde dehydrogenases (ALDH) 1A1/2 activity. At diagnosis, ROS level and ALDH1A1/2 activity in AML patients BM are correlated with the different ELN 2022 prognostic groups and overall survival (OS). A significant lower ALDH1A1/2 activity in BM was observed in the favourable ELN2022 subgroup compared to the intermediate and adverse group (p < 0.01). In the same way, the ROS levels were significantly lower in the favourable ELN 2022 subgroup compared to the intermediate group (p < 0.0001) and adverse group (p < 0.0002). ROShigh AML patients had a significantly lower median overall survival (OS) (8.2 months) than ROSlow patients (24.6 months) (p = 0.0368). After first-line therapy, a significant increase of ROS level (p = 0.015) and ALDH1A1/2 activity (0 = 0.0273) in leukaemic blasts was observed, especially in the refractory ones. ABD-3001, a competitive and irreversible inhibitor of ALDHs 1 and 3, can in vitro inhibit the proliferation of patient-derived leukaemic cells in accordance with redox balance. In multivariate analysis, ROS level was the most significant (p < 0.05) and the strongest predictive factor for the sensitivity of cells to ABD-3001. The safety profile of ABD-3001 is currently being assessed through the first inhuman multicenter phase 1 clinical trial "ODYSSEY" (NCT05601726) for patients with relapsed AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Reactive Oxygen Species/metabolism
- Prognosis
- Male
- Biomarkers, Tumor/metabolism
- Middle Aged
- Female
- Aldehyde Dehydrogenase 1 Family/metabolism
- Adult
- Aged
- Retinal Dehydrogenase/metabolism
- Aged, 80 and over
- Young Adult
- Aldehyde Dehydrogenase, Mitochondrial/metabolism
- Aldehyde Dehydrogenase, Mitochondrial/genetics
- Aldehyde Dehydrogenase/metabolism
Collapse
Affiliation(s)
- G. Venton
- APHM, Hôpital de la Conception, Service d'Hématologie et Thérapie CellulaireMarseilleFrance
- TAGC‐Theories and Approaches of Genomic ComplexityAix Marseille UniversityMarseilleFrance
| | - J. Colle
- APHM, Hôpital de la Conception, Service d'Hématologie et Thérapie CellulaireMarseilleFrance
- TAGC‐Theories and Approaches of Genomic ComplexityAix Marseille UniversityMarseilleFrance
| | - A. Tichadou
- APHM, Hôpital de la Conception, Service d'Hématologie et Thérapie CellulaireMarseilleFrance
- TAGC‐Theories and Approaches of Genomic ComplexityAix Marseille UniversityMarseilleFrance
| | - J. Quessada
- APHM, Hôpital La Timone, Laboratoire d'HématologieMarseilleFrance
| | | | | | | | - L. De Lassus
- APHM, Hôpital de la Conception, Service d'Hématologie et Thérapie CellulaireMarseilleFrance
| | - M. Loosveld
- APHM, Hôpital La Timone, Laboratoire d'HématologieMarseilleFrance
- CRCMInserm UMR1068, CNRS UMR7258, Aix Marseille Université U105, Institut Paoli CalmettesMarseilleFrance
| | - I. Arnoux
- APHM, Hôpital La Timone, Laboratoire d'HématologieMarseilleFrance
| | - N. Abbou
- APHM, Hopital La Timone, Service d'Oncobiologie, Plateforme M2GM, Hopital de la TimoneMarseilleFrance
- Aix‐Marseille Univ, INSERM, INRAE, C2VN, Laboratory of Haematology, CRB Assistance Publique‐Hôpitaux de Marseille, HemoVasc (CRB AP‐HM HemoVasc)MarseilleFrance
| | | | | | - R. Costello
- APHM, Hôpital de la Conception, Service d'Hématologie et Thérapie CellulaireMarseilleFrance
- TAGC‐Theories and Approaches of Genomic ComplexityAix Marseille UniversityMarseilleFrance
| |
Collapse
|
8
|
Kim JH, Lee J, Lee KW, Xiong H, Li M, Kim JS. Trapped in Cells: A Selective Accumulation Approach for Type-I Photodynamic Ablation of Cancer Stem-like Cells. JACS AU 2024; 4:3657-3667. [PMID: 39328753 PMCID: PMC11423316 DOI: 10.1021/jacsau.4c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024]
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme responsible for converting aldehyde functional groups into carboxylate metabolites. Elevated ALDH activity is a characteristic feature of cancer stem-like cells (CSCs). As a novel approach to target the CSC trait of overexpressing ALDH, we aimed to utilize ALDH activity for the selective accumulation of a photosensitizer in ALDHHigh CSCs. A novel ALDH substrate photosensitizer, SCHO, with thionylated coumarin and N-ethyl-4-(aminomethyl)benzaldehyde was developed to achieve this goal. Our study demonstrated the efficient metabolism of the aldehyde unit of SCHO into carboxylate, leading to its accumulation in ALDHHigh MDA-MB-231 cells. Importantly, we established the selectivity of SCHO as an ALDHHigh cell photosensitizer as it is not a substrate for ABC transporters. SCHO-based photodynamic therapy triggers apoptosis and pyroptosis in MDA-MB-231 cells and further reduces the characteristics of CSCs. Our study presents a novel strategy to target CSCs by exploiting their cellular metabolism to enhance photosensitizer accumulation, highlighting the potential of photodynamic therapy as a powerful tool for eliminating ALDHHigh CSCs.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- Department
of Chemical and Systems Biology, Chem-H
and Stanford Cancer Institute, Stanford School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Jieun Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Kyung-Woo Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Hao Xiong
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
9
|
Huang L, Liu M, Tang J, Gong Z, Li Z, Yang Y, Zhang M. The role of ALDH2 rs671 polymorphism and C-reactive protein in the phenotypes of male ALS patients. Front Neurosci 2024; 18:1397991. [PMID: 39290715 PMCID: PMC11405379 DOI: 10.3389/fnins.2024.1397991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024] Open
Abstract
Background The aldehyde dehydrogenase 2 (ALDH2) rs671 (A) allele has been implicated in neurodegeneration, potentially through oxidative and inflammatory pathways. The study aims to investigate the effects of the ALDH2 rs671 (A) allele and high sensitivity C-reactive protein (hs-CRP) on the clinical phenotypes of amyotrophic lateral sclerosis (ALS) in male and female patients. Methods Clinical data and ALDH2 rs671 genotype of 143 ALS patients, including 85 males and 58 females, were collected from January 2018 to December 2022. All patients underwent assessment using the Chinese version of the Edinburgh Cognitive and Behavioral ALS Screen (ECAS). Complete blood count and metabolic profiles were measured. Clinical and laboratory parameters were compared between carriers and non-carriers of the rs671 (A) allele in males and females, respectively. The significant parameters and rs671 (A) Allele were included in multivariate linear regression models to identify potential contributors to motor and cognitive impairment. Mediation analysis was employed to evaluate any mediation effects. Results Male patients carrying rs671 (A) allele exhibited higher levels of hs-CRP than non-carriers (1.70 mg/L vs. 0.50 mg/L, p = 0.006). The rs671 (A) allele was identified as an independent risk factor for faster disease progression only in male patients (β = 0.274, 95% CI = 0.048-0.499, p = 0.018). The effect of the rs671 (A) allele on the executive function in male patients was fully mediated by hs-CRP (Indirect effect = -1.790, 95% CI = -4.555--0.225). No effects of the rs671 (A) allele or hs-CRP were observed in female ALS patients. The effects of the ALDH2 rs671 (A) allele and the mediating role of hs-CRP in male patients remained significant in the sensitivity analyses. Conclusion The ALDH2 rs671 (A) allele contributed to faster disease progression and hs-CRP mediated cognitive impairment in male ALS patients.
Collapse
Affiliation(s)
- Lifang Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mao Liu
- Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
| | - Jiahui Tang
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhenxiang Gong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zehui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Yang
- Department of Neurology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Neurology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
10
|
Matsumoto A. Infection burden and ALDH2 rs671: East Asian genetic diversity. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1810-1811. [PMID: 38837888 DOI: 10.1111/acer.15380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024]
Affiliation(s)
- Akiko Matsumoto
- Department of Social and Environmental Medicine, Saga University, Saga, Japan
| |
Collapse
|
11
|
Ray B, Rungratanawanich W, LeFort KR, Chidambaram SB, Song BJ. Mitochondrial Aldehyde Dehydrogenase 2 (ALDH2) Protects against Binge Alcohol-Mediated Gut and Brain Injury. Cells 2024; 13:927. [PMID: 38891060 PMCID: PMC11171926 DOI: 10.3390/cells13110927] [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: 04/17/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Mitochondrial aldehyde dehydrogenase-2 (ALDH2) metabolizes acetaldehyde to acetate. People with ALDH2 deficiency and Aldh2-knockout (KO) mice are more susceptible to alcohol-induced tissue damage. However, the underlying mechanisms behind ALDH2-related gut-associated brain damage remain unclear. Age-matched young female Aldh2-KO and C57BL/6J wild-type (WT) mice were gavaged with binge alcohol (4 g/kg/dose, three doses) or dextrose (control) at 12 h intervals. Tissues and sera were collected 1 h after the last ethanol dose and evaluated by histological and biochemical analyses of the gut and hippocampus and their extracts. For the mechanistic study, mouse neuroblast Neuro2A cells were exposed to ethanol with or without an Aldh2 inhibitor (Daidzin). Binge alcohol decreased intestinal tight/adherens junction proteins but increased oxidative stress-mediated post-translational modifications (PTMs) and enterocyte apoptosis, leading to elevated gut leakiness and endotoxemia in Aldh2-KO mice compared to corresponding WT mice. Alcohol-exposed Aldh2-KO mice also showed higher levels of hippocampal brain injury, oxidative stress-related PTMs, and neuronal apoptosis than the WT mice. Additionally, alcohol exposure reduced Neuro2A cell viability with elevated oxidative stress-related PTMs and apoptosis, all of which were exacerbated by Aldh2 inhibition. Our results show for the first time that ALDH2 plays a protective role in binge alcohol-induced brain injury partly through the gut-brain axis, suggesting that ALDH2 is a potential target for attenuating alcohol-induced tissue injury.
Collapse
Affiliation(s)
- Bipul Ray
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA; (B.R.); (W.R.); (K.R.L.)
| | - Wiramon Rungratanawanich
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA; (B.R.); (W.R.); (K.R.L.)
| | - Karli R. LeFort
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA; (B.R.); (W.R.); (K.R.L.)
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, and Center for Experimental Pharmacology and Toxicology, JSS Academy of Higher Education & Research, Mysuru 570015, India;
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA; (B.R.); (W.R.); (K.R.L.)
| |
Collapse
|
12
|
Tang J, Alford A, Leung G, Tully M, Shi R. Neuroprotection by acrolein sequestration through exogenously applied scavengers and endogenous enzymatic enabling strategies in mouse EAE model. Sci Rep 2024; 14:6027. [PMID: 38472318 PMCID: PMC10933361 DOI: 10.1038/s41598-024-56035-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
We have previously shown that the pro-oxidative aldehyde acrolein is a critical factor in MS pathology. In this study, we found that the acrolein scavenger hydralazine (HZ), when applied from the day of induction, can suppress acrolein and alleviate motor and sensory deficits in a mouse experimental autoimmune encephalomyelitis (EAE) model. Furthermore, we also demonstrated that HZ can alleviate motor deficits when applied after the emergence of MS symptoms, making potential anti-acrolein treatment a more clinically relevant strategy. In addition, HZ can reduce both acrolein and MPO, suggesting a connection between acrolein and inflammation. We also found that in addition to HZ, phenelzine (PZ), a structurally distinct acrolein scavenger, can mitigate motor deficits in EAE when applied from the day of induction. This suggests that the likely chief factor of neuroprotection offered by these two structurally distinct acrolein scavengers in EAE is their common feature of acrolein neutralization. Finally, up-and-down regulation of the function of aldehyde dehydrogenase 2 (ALDH2) in EAE mice using either a pharmacological or genetic strategy led to correspondent motor and sensory changes. This data indicates a potential key role of ALDH2 in influencing acrolein levels, oxidative stress, inflammation, and behavior in EAE. These findings further consolidate the critical role of aldehydes in the pathology of EAE and its mechanisms of regulation. This is expected to reinforce and expand the possible therapeutic targets of anti-aldehyde treatment to achieve neuroprotection through both endogenous and exogenous manners.
Collapse
Affiliation(s)
- Jonathan Tang
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Anna Alford
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Gary Leung
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Melissa Tully
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
- MSTP Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
13
|
Tokiya M, Hashimoto M, Fukuda K, Kawamoto K, Akao C, Tsuji M, Yakushiji Y, Koike H, Matsumoto A. Asian flush gene variant increases mild cognitive impairment risk: a cross-sectional study of the Yoshinogari Brain MRI Checkup Cohort. Environ Health Prev Med 2024; 29:55. [PMID: 39401906 PMCID: PMC11473384 DOI: 10.1265/ehpm.24-00214] [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: 07/15/2024] [Accepted: 09/15/2024] [Indexed: 10/17/2024] Open
Abstract
BACKGROUND The East Asian-specific genetic diversity, the rs671 variant of aldehyde dehydrogenase 2, causes the "Asian flush" phenomenon following alcohol consumption, resulting in an alcohol avoidance phenotype. The variant is suggested as a risk factor for Alzheimer's disease; however, its association with mild cognitive impairment (MCI), an effective target for secondary prevention of dementia, remains unclear. METHOD This cross-sectional study examined 430 individuals aged 60-80 years (251 women) without overt cognitive impairment in Yoshinogari, Japan. The effect of the rs671 variant on MCI, defined by scores <26 or <25 on the Japanese version of the Montreal Cognitive Assessment, was evaluated using multivariate logistic regression. RESULTS The models included APOEε4, sex, age, education, history of habitual drinking, Brinkman index, hypertension, diabetes, and subclinical magnetic resonance imaging findings and consistently estimated the risk of the rs671 variant. Subsequently, stratified analyses by history of habitual drinking were performed based on an interactive effect between rs671 and alcohol consumption, and the rs671 variant significantly influenced MCI in participants who did not drink habitually, with odds ratios ranging from 1.9 to 2.1 before and after adjusting for covariates, suggesting an association independent of hippocampal atrophy and small vessel dysfunction. Conversely, no such association with the rs671 variant was observed in participants with a history of habitual alcohol use. Instead, hippocampal atrophy and silent infarcts were associated with MCI. CONCLUSIONS This is the first study to demonstrate an association between the rs671 variant and MCI morbidity. The findings highlight the need for race-specific preventive strategies and suggest potential unrecognized mechanisms in dementia development.
Collapse
Affiliation(s)
- Mikiko Tokiya
- Department of Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Manabu Hashimoto
- National Hospital Organization Hizen Psychiatric Medical Center, 160 Mitsu, Yoshinogari-machi, Kanzaki-gun, Saga 842-0192, Japan
| | - Kenji Fukuda
- Department of Cerebrovascular Disease, St. Mary’s Hospital, 422 Tsubukuhonmachi, Kurume, Fukuoka 830-8543, Japan
| | - Kazuhiro Kawamoto
- Department of Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Chiho Akao
- Department of Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Mariko Tsuji
- National Hospital Organization Hizen Psychiatric Medical Center, 160 Mitsu, Yoshinogari-machi, Kanzaki-gun, Saga 842-0192, Japan
| | - Yusuke Yakushiji
- Department of Neurology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka-fu 573-1010, Japan
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Haruki Koike
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Akiko Matsumoto
- Department of Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
14
|
Takashima S, Tokiya M, Izui K, Miyamoto H, Matsumoto A. Asian flush is a potential protective factor against COVID-19: a web-based retrospective survey in Japan. Environ Health Prev Med 2024; 29:14. [PMID: 38462476 PMCID: PMC10937249 DOI: 10.1265/ehpm.23-00361] [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: 12/14/2023] [Accepted: 02/10/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), first reported in December 2019, spread worldwide in a short period, resulting in numerous cases and associated deaths; however, the toll was relatively low in East Asia. A genetic polymorphism unique to East Asians, Aldehyde dehydrogenase 2 rs671, has been reported to confer protection against infections. METHOD We retrospectively investigated the association between the surrogate marker of the rs671 variant, the skin flushing phenomenon after alcohol consumption, and the timing of COVID-19 incidence using a web-based survey tool to test any protective effects of rs671 against COVID-19. RESULTS A total of 807 valid responses were received from 362 non-flushers and 445 flushers. During the 42 months, from 12/1/2019 to 5/31/2023, 40.6% of non-flushers and 35.7% of flushers experienced COVID-19. Flushers tended to have a later onset (Spearman's partial rank correlation test, p = 0.057, adjusted for sex and age). Similarly, 2.5% of non-flushers and 0.5% of flushers were hospitalized because of COVID-19. Survival analysis estimated lower risks of COVID-19 and associated hospitalization among flushers (p = 0.03 and <0.01, respectively; generalized Wilcoxon test). With the Cox proportional hazards model covering 21 months till 8/31/2021, when approximately half of the Japanese population had received two doses of COVID-19 vaccine, the hazard ratio (95% confidence interval) of COVID-19 incidence was estimated to be 0.21 (0.10-0.46) for flusher versus non-flusher, with adjustment for sex, age, steroid use, and area of residence. CONCLUSIONS Our study suggests an association between the flushing phenomenon after drinking and a decreased risk of COVID-19 morbidity and hospitalization, suggesting that the rs671 variant is a protective factor. This study provides valuable information for infection control and helps understand the unique constitutional diversity of East Asians.
Collapse
Affiliation(s)
- Satoshi Takashima
- Department of Social and Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
- Plant Products Safety Division, Food Safety and Consumer Affairs Bureau, Ministry of Agriculture, Forestry and Fisheries, 1-2-1 Kasumigaseki, Chiyodaku, Tokyo 100-8950, Japan
| | - Mikiko Tokiya
- Department of Social and Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Katsura Izui
- Graduate School of Biostudies, Kyoto University, Oiwake-cho, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hiroshi Miyamoto
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Akiko Matsumoto
- Department of Social and Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
15
|
Sołtysiak M, Paplińska-Goryca M, Misiukiewicz-Stępień P, Wójtowicz P, Dutkiewicz M, Zegrocka-Stendel O, Sikorska M, Dymkowska D, Turos-Korgul L, Krenke R, Koziak K. β-escin activates ALDH and prevents cigarette smoke-induced cell death. Biomed Pharmacother 2024; 170:115924. [PMID: 38016364 DOI: 10.1016/j.biopha.2023.115924] [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: 08/07/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND The tobacco use is one of the biggest public health threats worldwide. Cigarette smoke contains over 7000 chemicals among other aldehydes, regarded as priority toxicants. β-escin (a mixture of triterpenoid saponins extracted from the Aesculus hippocastanum. L) is a potent activator of aldehyde dehydrogenase (ALDH) - an enzyme catalyzing oxidation of aldehydes to non-toxic carboxylic acids. PURPOSE The aim of this study was to evaluate the effect of β-escin on ALDH activity, ALDH isoforms mRNA expression and cytotoxicity in nasal epithelial cells exposed to cigarette smoke extract (CSE). METHODS Nasal epithelial cells from healthy non-smokers were treated with β-escin (1 µM) and exposed to 5% CSE. After 6- or 24-hours of stimulation cell viability, DNA damage, ALDH activity and mRNA expression of ALDH isoforms were examined. RESULTS 24 h β-escin stimulation revised CSE induced cytotoxicity and DNA damage. Cells cultured with β-escin or exposed to CSE responded with strong increase in ALDH activity. This effect was more pronounced in cultures treated with combination of β-escin and CSE. The strongest stimulatory effect on ALDH isoform mRNA expression was observed in cells cultured simultaneously with β-escin and CSE: at 6 h for ALDH1A1 and ALDH3A1, and at 24 h for ALDH1A3, ALDH3A2, ALDH3B1, and ALDH18A1. Combined β-escin and CSE treatment prevented the CSE-induced inhibition of ALDH2 expression at 24 h. CONCLUSIONS β-escin is an effective ALDH stimulatory and cytoprotective agent and might be useful in the prevention or supportive treatment of tobacco smoke-related diseases.
Collapse
Affiliation(s)
- Malwina Sołtysiak
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Magdalena Paplińska-Goryca
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Paulina Misiukiewicz-Stępień
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Paulina Wójtowicz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Małgorzata Dutkiewicz
- Department of Biochemistry and Nutrition, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
| | - Oliwia Zegrocka-Stendel
- Department of Biochemistry and Nutrition, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
| | - Maria Sikorska
- Department of Biochemistry and Nutrition, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
| | - Dorota Dymkowska
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteura 3, 02-093 Warsaw, Poland
| | - Laura Turos-Korgul
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteura 3, 02-093 Warsaw, Poland
| | - Rafał Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland
| | - Katarzyna Koziak
- Department of Biochemistry and Nutrition, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland.
| |
Collapse
|
16
|
Gerber TS, Witzel HR, Weinmann A, Bartsch F, Schindeldecker M, Galle PR, Lang H, Roth W, Ridder DA, Straub BK. Reduced Lipid Peroxidation Predicts Unfavorable Prognosis in Hepatocellular Carcinoma, but Not Intrahepatic Cholangiocarcinoma. Biomedicines 2023; 11:2471. [PMID: 37760911 PMCID: PMC10525544 DOI: 10.3390/biomedicines11092471] [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: 07/28/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Primary liver cancer, including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), remains a significant contributor to cancer-related mortality worldwide. Oxidative stress and lipid peroxidation play a key role in chronic liver diseases and have been shown to be pivotal for tumor initiation and progression. 4-hydroxy-nonenal (4-HNE), one of the major mediators of oxidative stress and a well-established biomarker for lipid peroxidation, can act as a signal transducer, inducing inflammation and exerting carcinogenic effects. However, the role of 4-HNE in primary liver cancer remains poorly explored. In this study, we investigated 4-HNE levels in 797 liver carcinomas, including 561 HCC and 236 iCCA, by immunohistochemistry. We then correlated 4-HNE levels with comprehensive clinical data and survival outcomes. In HCC, lower expression levels of 4-HNE were associated with vascular invasion, a high tumor grade, a macrotrabecular-massive HCC subtype, and poor overall survival. Concerning iCCA, large duct iCCA showed significantly higher 4-HNE levels when compared to small duct iCCA. Yet, in iCCA, 4-HNE levels did not correlate with known prognostic parameters or survival outcomes. To conclude, in HCC but not in iCCA, low amounts of 4-HNE predict unfavorable survival outcomes and are associated with aggressive tumor behavior. These findings provide insights into the role of 4-HNE in liver cancer progression and may enable novel therapeutic strategies.
Collapse
Affiliation(s)
- Tiemo Sven Gerber
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.S.G.); (H.R.W.); (M.S.); (W.R.); (D.A.R.)
| | - Hagen Roland Witzel
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.S.G.); (H.R.W.); (M.S.); (W.R.); (D.A.R.)
| | - Arndt Weinmann
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (A.W.); (P.R.G.)
| | - Fabian Bartsch
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (F.B.); (H.L.)
| | - Mario Schindeldecker
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.S.G.); (H.R.W.); (M.S.); (W.R.); (D.A.R.)
- Tissue Biobank, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Peter R. Galle
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (A.W.); (P.R.G.)
| | - Hauke Lang
- Department of General, Visceral and Transplant Surgery, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (F.B.); (H.L.)
| | - Wilfried Roth
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.S.G.); (H.R.W.); (M.S.); (W.R.); (D.A.R.)
| | - Dirk Andreas Ridder
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.S.G.); (H.R.W.); (M.S.); (W.R.); (D.A.R.)
| | - Beate Katharina Straub
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.S.G.); (H.R.W.); (M.S.); (W.R.); (D.A.R.)
| |
Collapse
|
17
|
Ye Y, Wu T, Liang F, Fan J, Song P, Li Y, Xie W, Huang X, Han P. Prognostic Significance of a Model Based on Acetaldehyde Dehydrogenase 2 Genetic Polymorphisms in Laryngeal Carcinoma. Otolaryngol Head Neck Surg 2023; 169:528-538. [PMID: 36758951 DOI: 10.1002/ohn.266] [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: 08/07/2022] [Revised: 11/28/2022] [Accepted: 12/03/2022] [Indexed: 02/11/2023]
Abstract
OBJECTIVE Because of the high costs associated with early-stage laryngeal carcinoma diagnosis and prognosis prediction, this study attempts to find valuable targets to establish a novel predictive model by focusing on the aldehyde dehydrogenase 2 (ALDH2) genotype and other peripheral blood markers. STUDY DESIGN Retrospective study. SETTING Tertiary comprehensive hospital. METHODS From January 2011 to January 2021, 362 cases of laryngeal carcinoma were included and divided into 2 groups in this retrospective analysis. Information on medical history, alcohol, and tobacco consumption habits, ALDH2 genotypes, and other peripheral blood markers was collected. Endpoints of the current study included disease-free survival and overall survival. A nomogram model for overall survival was established and evaluated using receiver operating characteristic (ROC) curves. RESULTS A total of 236 patients were included in the training cohort, and the other 126 were included in the validation cohort. The median follow-up of the patients was 9.6 years (interquartile range: 7.5-12.5 years). Peripheral fibrinogen, hemoglobin, and ALDH2 genotypes were significantly associated with an increase in laryngeal carcinoma mortality rate on Kaplan-Meier curves. The ROC curve showed that the effectiveness of overall survival prediction by the nomogram model was better than that of traditional clinical staging. CONCLUSION A prognostic nomogram of laryngeal carcinoma patients involving ALDH2 and peripheral blood markers and T and N stages was constructed and validated.
Collapse
Affiliation(s)
- Yuchu Ye
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Taowei Wu
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Faya Liang
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Jianming Fan
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Pan Song
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Yixin Li
- Department of Otolaryngology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wenqian Xie
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Xiaoming Huang
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Ping Han
- Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| |
Collapse
|
18
|
Berry SB, Espich S, Thuong NTT, Chang X, Dorajoo R, Khor CC, Heng CK, Yuan JM, Fox D, Anaya-Sanchez A, Tenney L, Chang CJ, Kotov DI, Vance RE, Dunstan SJ, Darwin KH, Stanley SA. Disruption of Aldehyde Dehydrogenase 2 protects against bacterial infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554661. [PMID: 37662190 PMCID: PMC10473740 DOI: 10.1101/2023.08.24.554661] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The ALDH2*2 (rs671) allele is one of the most common genetic mutations in humans, yet the positive evolutionary selective pressure to maintain this mutation is unknown, despite its association with adverse health outcomes. ALDH2 is responsible for the detoxification of metabolically produced aldehydes, including lipid-peroxidation end products derived from inflammation. Here, we demonstrate that host-derived aldehydes 4-hydroxynonenal (4HNE), malondialdehyde (MDA), and formaldehyde (FA), all of which are metabolized by ALDH2, are directly toxic to the bacterial pathogens Mycobacterium tuberculosis and Francisella tularensis at physiological levels. We find that Aldh2 expression in macrophages is decreased upon immune stimulation, and that bone marrow-derived macrophages from Aldh2 -/- mice contain elevated aldehydes relative to wild-type mice. Macrophages deficient for Aldh2 exhibited enhanced control of Francisella infection. Finally , mice lacking Aldh2 demonstrated increased resistance to pulmonary infection by M. tuberculosis , including in a hypersusceptible model of tuberculosis, and were also resistant to Francisella infection. We hypothesize that the absence of ALDH2 contributes to the host's ability to control infection by pathogens such as M. tuberculosis and F. tularensis , and that host-derived aldehydes act as antimicrobial factors during intracellular bacterial infections. One sentence summary Aldehydes produced by host cells contribute to the control of bacterial infections.
Collapse
|
19
|
Karan BM, Little K, Augustine J, Stitt AW, Curtis TM. Aldehyde Dehydrogenase and Aldo-Keto Reductase Enzymes: Basic Concepts and Emerging Roles in Diabetic Retinopathy. Antioxidants (Basel) 2023; 12:1466. [PMID: 37508004 PMCID: PMC10376360 DOI: 10.3390/antiox12071466] [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: 06/27/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetic retinopathy (DR) is a complication of diabetes mellitus that can lead to vision loss and blindness. It is driven by various biochemical processes and molecular mechanisms, including lipid peroxidation and disrupted aldehyde metabolism, which contributes to retinal tissue damage and the progression of the disease. The elimination and processing of aldehydes in the retina rely on the crucial role played by aldehyde dehydrogenase (ALDH) and aldo-keto reductase (AKR) enzymes. This review article investigates the impact of oxidative stress, lipid-derived aldehydes, and advanced lipoxidation end products (ALEs) on the advancement of DR. It also provides an overview of the ALDH and AKR enzymes expressed in the retina, emphasizing their growing importance in DR. Understanding the relationship between aldehyde metabolism and DR could guide innovative therapeutic strategies to protect the retina and preserve vision in diabetic patients. This review, therefore, also explores various approaches, such as gene therapy and pharmacological compounds that have the potential to augment the expression and activity of ALDH and AKR enzymes, underscoring their potential as effective treatment options for DR.
Collapse
Affiliation(s)
- Burak Mugdat Karan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Karis Little
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Tim M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| |
Collapse
|
20
|
Lee J, Roh JL. Targeting Nrf2 for ferroptosis-based therapy: Implications for overcoming ferroptosis evasion and therapy resistance in cancer. Biochim Biophys Acta Mol Basis Dis 2023:166788. [PMID: 37302427 DOI: 10.1016/j.bbadis.2023.166788] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/10/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Ferroptosis is a newly discovered form of programmed cell death caused by redox-active iron-mediated lipid peroxidation. Ferroptosis exhibits a unique morphological phenotype resulting from oxidative damage to membrane lipids. Ferroptosis induction has been shown to be effective in treating human cancers that rely on lipid peroxidation repair pathways. Nuclear factor erythroid 2-related factor 2 (Nrf2) can control the regulatory pathways of ferroptosis, which involve genes associated with glutathione biosynthesis, antioxidant responses, and lipid and iron metabolism. Resistant cancer cells often utilize Nrf2 stabilization by Keap1 inactivation or other somatic alterations in the genes from the Nrf2 pathway, which can confer resistance to ferroptosis induction and other therapies. However, pharmacological inactivation of the Nrf2 pathway can sensitize cancer cells to ferroptosis induction. Inducing lipid peroxidation and ferroptosis through regulating the Nrf2 pathway is a promising strategy for enhancing the anticancer effects of chemotherapy and radiation therapy in therapy-resistant human cancers. Despite promising preliminary studies, clinical trials in human cancer therapy have not yet been realized. A deeper understanding of their exact processes and efficacies in various cancers remains unsolved. Therefore, this article aims to summarize the regulatory mechanisms of ferroptosis, their modulation by Nrf2, and the potential of targeting Nrf2 for ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
21
|
Dancik GM, Varisli L, Vlahopoulos SA. The Molecular Context of Oxidant Stress Response in Cancer Establishes ALDH1A1 as a Critical Target: What This Means for Acute Myeloid Leukemia. Int J Mol Sci 2023; 24:ijms24119372. [PMID: 37298333 DOI: 10.3390/ijms24119372] [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: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The protein family of aldehyde dehydrogenases (ALDH) encompasses nineteen members. The ALDH1 subfamily consists of enzymes with similar activity, having the capacity to neutralize lipid peroxidation products and to generate retinoic acid; however, only ALDH1A1 emerges as a significant risk factor in acute myeloid leukemia. Not only is the gene ALDH1A1 on average significantly overexpressed in the poor prognosis group at the RNA level, but its protein product, ALDH1A1 protects acute myeloid leukemia cells from lipid peroxidation byproducts. This capacity to protect cells can be ascribed to the stability of the enzyme under conditions of oxidant stress. The capacity to protect cells is evident both in vitro, as well as in mouse xenografts of those cells, shielding cells effectively from a number of potent antineoplastic agents. However, the role of ALDH1A1 in acute myeloid leukemia has been unclear in the past due to evidence that normal cells often have higher aldehyde dehydrogenase activity than leukemic cells. This being true, ALDH1A1 RNA expression is significantly associated with poor prognosis. It is hence imperative that ALDH1A1 is methodically targeted, particularly for the acute myeloid leukemia patients of the poor prognosis risk group that overexpress ALDH1A1 RNA.
Collapse
Affiliation(s)
- Garrett M Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Spiros A Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
| |
Collapse
|
22
|
Camilleri AE, Cung M, Hart FM, Pagovich OE, Crystal RG, Greenblatt MB, Stiles KM. Gene Therapy to Treat Osteopenia Associated With Chronic Ethanol Consumption and Aldehyde Dehydrogenase 2 Deficiency. JBMR Plus 2023; 7:e10723. [PMID: 37065630 PMCID: PMC10097638 DOI: 10.1002/jbm4.10723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 01/27/2023] Open
Abstract
Aldehyde dehydrogenase 2 (ALDH2) deficiency affects 35% to 45% of East Asians and 8% of the world population. ALDH2 is the second enzyme in the ethanol metabolism pathway. The common genetic variant ALDH2*2 allele has a glutamic acid-to-lysine substitution at position 487 (E487K) that reduces the enzyme activity, resulting in an accumulation of acetaldehyde after ethanol consumption. The ALDH2*2 allele is associated with increased risk of osteoporosis and hip fracture. Our prior study showed that administration of an adeno-associated virus (AAV) serotype rh.10 gene transfer vector expressing the human ALDH2 cDNA (AAVrh.10hALDH2) before initiation of ethanol consumption prevented bone loss in ALDH2-deficient homozygous knockin mice carrying the E487K mutation (Aldh2 E487K+/+). We hypothesized that AAVrh.10hALDH2 administration after establishment of osteopenia would be able to reverse bone loss due to ALDH2 deficiency and chronic ethanol consumption. To test this hypothesis, male and female Aldh2 E487K+/+ mice (n = 6) were given ethanol in the drinking water for 6 weeks to establish osteopenia and then administered AAVrh.10hALDH2 (1011 genome copies). Mice were evaluated for an additional 12 weeks. AAVrh.10hALDH2 administration after osteopenia was established corrected weight loss and locomotion phenotypes and, importantly, increased midshaft femur cortical bone thickness, the most important component of bone in the resistance to fractures, and showed a trend toward increased trabecular bone volume. AAVrh.10hALDH2 is a promising therapeutic for osteoporosis in ALDH2-deficient individuals. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Anna E Camilleri
- Department of Genetic MedicineWeill Cornell Medical CollegeNew YorkNYUSA
| | - Michelle Cung
- Pathology and Laboratory MedicineWeill Cornell Medical CollegeNew YorkNYUSA
| | - Fiona M Hart
- Department of Genetic MedicineWeill Cornell Medical CollegeNew YorkNYUSA
| | - Odelya E Pagovich
- Department of Genetic MedicineWeill Cornell Medical CollegeNew YorkNYUSA
| | - Ronald G Crystal
- Department of Genetic MedicineWeill Cornell Medical CollegeNew YorkNYUSA
| | - Matthew B Greenblatt
- Pathology and Laboratory MedicineWeill Cornell Medical CollegeNew YorkNYUSA
- Research DivisionHospital for Special SurgeryNew YorkNYUSA
| | - Katie M Stiles
- Department of Genetic MedicineWeill Cornell Medical CollegeNew YorkNYUSA
| |
Collapse
|
23
|
Tran TO, Vo TH, Lam LHT, Le NQK. ALDH2 as a potential stem cell-related biomarker in lung adenocarcinoma: Comprehensive multi-omics analysis. Comput Struct Biotechnol J 2023; 21:1921-1929. [PMID: 36936815 PMCID: PMC10018390 DOI: 10.1016/j.csbj.2023.02.045] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/06/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the most prevalent lung cancer and one of the leading causes of death. Previous research found a link between LUAD and Aldehyde Dehydrogenase 2 (ALDH2), a member of aldehyde dehydrogenase gene (ALDH) superfamily. In this study, we identified additional useful prognostic markers for early LUAD identification and targeting LUAD therapy by analyzing the expression level, epigenetic mechanism, and signaling activities of ALDH2 in LUAD patients. The obtained results demonstrated that ALDH2 gene and protein expression significantly downregulated in LUAD patient samples. Furthermore, The American Joint Committee on Cancer (AJCC) reported that diminished ALDH2 expression was closely linked to worse overall survival (OS) in different stages of LUAD. Considerably, ALDH2 showed aberrant DNA methylation status in LUAD cancer. ALDH2 was found to be downregulated in the proteomic expression profile of several cell biology signaling pathways, particularly stem cell-related pathways. Finally, the relationship of ALDH2 activity with stem cell-related factors and immune system were reported. In conclusion, the downregulation of ALDH2, abnormal DNA methylation, and the consequent deficit of stemness signaling pathways are relevant prognostic and therapeutic markers in LUAD.
Collapse
Key Words
- 4-HNE, 4-Hydroxynonenal
- AJCC, American Joint Committee On Cancer
- ALDH, Aldehyde Dehydrogenase
- Aldehyde Dehydrogenase 2
- CGI, Cpg Island
- CPTAC, Clinical Proteomic Tumor Analysis Consortium
- CSCs, Cancer Stem Cells
- Cancer stem cells
- DNA methylation
- Gene expression
- IHC, Immunohistochemical
- LCSCs, Liver Cancer Stem Cells
- LUAD, Lung Adenocarcinoma
- Lung adenocarcinoma
- MAPK, Mitogen-Activated Protein Kinase
- MDA, Malondialdehyde
- NSCLC, Non-Small Cell Lung Cancer
- OS, Overall Survival
- Protein expression
- ROS, Reactive Oxygen Species
- SCLC, Small Cell Lung Cancer
- Survival analysis
- TCGA, The Cancer Genome Atlas
- TMT, Tandem Mass Tags
- TNM, Tumor-Node-Metastasis
- UICC, International Union For Cancer Control
- XRCC1, X-Ray Repair Cross-Complementing Protein 1
Collapse
Affiliation(s)
- Thi-Oanh Tran
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Hematology and Blood Transfusion Center, Bach Mai Hospital, No.78, Giai Phong street, Hanoi, Viet Nam
| | - Thanh Hoa Vo
- Department of Science, School of Science and Computing, South East Technological University, Waterford X91 K0EK, Ireland
- Pharmaceutical and Molecular Biotechnology Research Center (PMBRC), South East Technological University, Waterford X91 K0EK, Ireland
| | - Luu Ho Thanh Lam
- Department of Pediatrics, Pham Ngoc Thach University of Medicine, Ho Chi Minh city, Viet Nam
- Children’s Hospital 1, Ho Chi Minh city, Viet Nam
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 106, Taiwan
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei 106, Taiwan
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Corresponding author at: Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 106, Taiwan.
| |
Collapse
|
24
|
Zhang H, Li Z, Zheng Y. Identifying the Therapeutic and Prognostic Role of the CD8+ T Cell-Related Gene ALDH2 in Head and Neck Squamous Cell Carcinoma. Cancer Inform 2022; 21:11769351221139252. [PMID: 36570380 PMCID: PMC9772952 DOI: 10.1177/11769351221139252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 10/28/2022] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSC) is a widely known malignancy which is usually diagnosed late and has a poor prognosis. This study focuses on finding a new gene linked with CD8+ T cell infiltration as a prognostic marker for patients with HNSC. Differential analysis of transcriptomic data was performed between HNSC and control tissues from TCGA and GEO database. The CD8+ T cell infiltration score was quantified using single-sample gene set enrichment analysis (ssGSEA). Weighted gene co-expression network analysis (WGCNA) algorithms were used to identify key modules associated with CD8+ T cell infiltration. Kaplan-Meier (K-M) survival analysis was used to compare overall survival (OS) between the 2 groups. Univariate and multivariate Cox analyses were used to assess independent prognostic markers. The results showed CD8+ T cell infiltration score was an independent favorable prognostic marker in HNSC. Differential analysis and WGCNA identified 93 differential gene related to high CD8+ T infiltration. Amog the 93 genes, ALDH2 was an independent favorable prognostic marker in HNSC. ALDH2 expression was found to be much lower in HNSC, and patients with low ALDH2 expression had higher T stage and N stage. The correlation analysis showed that ALDH2 was linked with immune cell infiltration in the tumor microenvironment of HNSC. Patients having increased expression of ALDH2 tend to be sensitive to immune checkpoint inhibitors (ICIs). In addition, we showed the relationship between ALDH2 expression and chemotherapeutic drug sensitivity. In conclusion, this study identified ALDH2 as a prognostic marker, associated with CD8+ T cell infiltration in HNSC.
Collapse
Affiliation(s)
- Hongmei Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhaozheng Li
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yan Zheng
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China,Yan Zheng, Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Xuefu Street 246, Harbin, 150086, China.
| |
Collapse
|
25
|
Gagestein B, von Hegedus JH, Kwekkeboom JC, Heijink M, Blomberg N, van der Wel T, Florea BI, van den Elst H, Wals K, Overkleeft HS, Giera M, Toes REM, Ioan-Facsinay A, van der Stelt M. Comparative Photoaffinity Profiling of Omega-3 Signaling Lipid Probes Reveals Prostaglandin Reductase 1 as a Metabolic Hub in Human Macrophages. J Am Chem Soc 2022; 144:18938-18947. [PMID: 36197299 PMCID: PMC9585591 DOI: 10.1021/jacs.2c06827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
The fish oil constituent
docosahexaenoic acid (DHA, 22:6
n-3) is
a signaling lipid with anti-inflammatory properties. The molecular
mechanisms underlying the biological effect of DHA are poorly understood.
Here, we report the design, synthesis, and application of a complementary
pair of bio-orthogonal, photoreactive probes based on the polyunsaturated
scaffold DHA and its oxidative metabolite 17-hydroxydocosahexaenoic
acid (17-HDHA). In these probes, an alkyne serves as a handle to introduce
a fluorescent reporter group or a biotin-affinity tag via copper(I)-catalyzed
azide-alkyne cycloaddition. This pair of chemical probes was used
to map specific targets of the omega-3 signaling lipids in primary
human macrophages. Prostaglandin reductase 1 (PTGR1) was identified
as an interaction partner that metabolizes 17-oxo-DHA, an oxidative
metabolite of 17-HDHA. 17-oxo-DHA reduced the formation of pro-inflammatory
lipids 5-HETE and LTB4 in human macrophages and neutrophils. Our results
demonstrate the potential of comparative photoaffinity protein profiling
for the discovery of metabolic enzymes of bioactive lipids and highlight
the power of chemical proteomics to uncover new biological insights.
Collapse
Affiliation(s)
- Berend Gagestein
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Johannes H von Hegedus
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Joanneke C Kwekkeboom
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Marieke Heijink
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Niek Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Bogdan I Florea
- Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Hans van den Elst
- Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Kim Wals
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Herman S Overkleeft
- Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Andreea Ioan-Facsinay
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| |
Collapse
|
26
|
Hu Q, Chen H, Shen C, Zhang B, Weng X, Sun X, Liu J, Dong Z, Hu K, Ge J, Sun A. Impact and potential mechanism of effects of chronic moderate alcohol consumption on cardiac function in aldehyde dehydrogenase 2 gene heterozygous mice. Alcohol Clin Exp Res 2022; 46:707-723. [PMID: 35315077 PMCID: PMC9321750 DOI: 10.1111/acer.14811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 12/01/2022]
Abstract
Background Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is a key enzyme in alcohol metabolism. The ALDH2*2 mutations are found in approximately 45% of East Asians, with 40% being heterozygous (HE) ALDH2*1/*2 and 5% homozygous (HO) ALDH2*2/*2. Studies have shown that HO mice lack cardioprotective effects induced by moderate alcohol consumption. However, the impact of moderate alcohol consumption on cardiac function in HE mice is unknown. Methods In this study, HO, HE, and wild‐type (WT) mice were subjected to a 6‐week moderate alcohol drinking protocol, following which myocardial tissue and cardiomyocytes of the mice were extracted. Results We found that moderate alcohol exposure did not increase mortality, myocardial fibrosis, apoptosis, or inflammation in HE mice, which differs from the effects observed in HO mice. After exposure to the 6‐week alcohol drinking protocol, there was impaired cardiac function, cardiomyocyte contractility, and intracellular Ca2+ homeostasis and mitochondrial function in both HE and HO mice as compared to WT mice. Moreover, these animals showed overt oxidative stress production and increased levels of the activated forms of calmodulin‐dependent protein kinase II (CaMKII) and ryanodine receptor type 2 (RYR2) phosphorylation protein. Conclusion We found that moderate alcohol exposure impaired cardiac function in HE mice, possibly by increasing reactive oxygen species (ROS)/CaMKII/RYR2‐mediated Ca2+ handling abnormalities. Hence, we advocate that people with ALDH2*1/*2 genotypes rigorously avoid alcohol consumption to prevent potential cardiovascular harm induced by moderate alcohol consumption.
Collapse
Affiliation(s)
- Qinfeng Hu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hang Chen
- Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, China.,Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Shen
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Beijian Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xiaolei Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jin Liu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Zhen Dong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Kai Hu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Aijun Sun
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
27
|
Wang W, Wang J, Liu S, Ren Y, Wang J, Liu S, Cui W, Jia L, Tang X, Yang J, Wu C, Wang L. An EHMT2/NFYA-ALDH2 signaling axis modulates the RAF pathway to regulate paclitaxel resistance in lung cancer. Mol Cancer 2022; 21:106. [PMID: 35477569 PMCID: PMC9044593 DOI: 10.1186/s12943-022-01579-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022] Open
Abstract
Background Lung cancer is a kind of malignancy with high morbidity and mortality worldwide. Paclitaxel (PTX) is the main treatment for non-small cell lung cancer (NSCLC), and resistance to PTX seriously affects the survival of patients. However, the underlying mechanism and potential reversing strategy need to be further explored. Methods We identified ALDH2 as a PTX resistance-related gene using gene microarray analysis. Subsequently, a series of functional analysis in cell lines, patient samples and xenograft models were performed to explore the functional role, clinical significance and the aberrant regulation mechanism of ALDH2 in PTX resistance of NSCLC. Furthermore, the pharmacological agents targeting ALDH2 and epigenetic enzyme were used to investigate the diverse reversing strategy against PTX resistance. Results Upregulation of ALDH2 expression is highly associated with resistance to PTX using in vitro and in vivo analyses of NSCLC cells along with clinicopathological analyses of NSCLC patients. ALDH2-overexpressing NSCLC cells exhibited significantly reduced PTX sensitivity and increased biological characteristics of malignancy in vitro and tumor growth and metastasis in vivo. EHMT2 (euchromatic histone lysine methyltransferase 2) inhibition and NFYA (nuclear transcription factor Y subunit alpha) overexpression had a cooperative effect on the regulation of ALDH2. Mechanistically, ALDH2 overexpression activated the RAS/RAF oncogenic pathway. NSCLC/PTX cells re-acquired sensitivity to PTX in vivo and in vitro when ALDH2 was inhibited by pharmacological agents, including the ALDH2 inhibitors Daidzin (DZN)/Disulfiram (DSF) and JIB04, which reverses the effect of EHMT2. Conclusion Our findings suggest that ALDH2 status can help predict patient response to PTX therapy and ALDH2 inhibition may be a promising strategy to overcome PTX resistance in the clinic. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-022-01579-9.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Jianmin Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Shuai Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Yong Ren
- Department of Pathology, General Hospital of Central Theater Command of People's Liberation Army, Wuhan, People's Republic of China
| | - Jingyu Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Sen Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Wei Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Lina Jia
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xing Tang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China. .,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, People's Republic of China. .,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, People's Republic of China.
| |
Collapse
|
28
|
Kusuma GD, Georgiou HM, Perkins AV, Abumaree MH, Brennecke SP, Kalionis B. Mesenchymal Stem/Stromal Cells and Their Role in Oxidative Stress Associated with Preeclampsia. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:115-127. [PMID: 35370491 PMCID: PMC8961706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Preeclampsia (PE) is a serious medically important disorder of human pregnancy, which features de novo pregnancy-induced hypertension and proteinuria. The severe form of PE can progress to eclampsia, a convulsive, life-threatening condition. When placental growth and perfusion are abnormal, the placenta experiences oxidative stress and subsequently secretes abnormal amounts of certain pro-angiogenic factors (eg, PlGF) as well as anti-angiogenic factors (eg, sFlt-1) that enter the maternal circulation. The net effect is damage to the maternal vascular endothelium, which subsequently manifests as the clinical features of PE. Other than delivery of the fetus and placenta, curative treatments for PE have not yet been forthcoming, which reflects the complexity of the clinical syndrome. A major source of reactive oxygen species that contributes to the widespread maternal vascular endothelium damage is the PE-affected decidua. The role of decidua-derived mesenchymal stem/stromal cells (MSC) in normotensive and pathological placenta development is poorly understood. The ability to respond to an environment of oxidative damage is a "universal property" of MSC but the biological mechanisms that MSC employ in response to oxidative stress are compromised in PE. In this review, we discuss how MSC respond to oxidative stress in normotensive and pathological conditions. We also consider the possibility of manipulating the oxidative stress response of abnormal MSC as a therapeutic strategy to treat preeclampsia.
Collapse
Affiliation(s)
- Gina D. Kusuma
- The University of Melbourne, Department of Obstetrics
and Gynaecology, Royal Women’s Hospital, Parkville, Victoria, Australia,Pregnancy Research Centre, Department of Maternal-Fetal
Medicine, Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Harry M. Georgiou
- The University of Melbourne, Department of Obstetrics
and Gynaecology, Royal Women’s Hospital, Parkville, Victoria, Australia,Pregnancy Research Centre, Department of Maternal-Fetal
Medicine, Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Anthony V. Perkins
- School of Medical Science, Menzies Health Institute
Queensland, Griffith University, Southport, Queensland, Australia
| | - Mohamed H. Abumaree
- Stem Cells and Regenerative Medicine Department, King
Abdullah International Medical Research Center, King Abdulaziz Medical City,
Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia,King Saud Bin Abdulaziz University for Health Sciences,
College of Science and Health Professions, King Abdulaziz Medical City, Ministry
of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Shaun P. Brennecke
- The University of Melbourne, Department of Obstetrics
and Gynaecology, Royal Women’s Hospital, Parkville, Victoria, Australia,Pregnancy Research Centre, Department of Maternal-Fetal
Medicine, Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Bill Kalionis
- The University of Melbourne, Department of Obstetrics
and Gynaecology, Royal Women’s Hospital, Parkville, Victoria, Australia,Pregnancy Research Centre, Department of Maternal-Fetal
Medicine, Royal Women’s Hospital, Parkville, Victoria, Australia,To whom all correspondence should be addressed:
Dr. Bill Kalionis, Department of Maternal-Fetal Medicine Pregnancy Research
Centre Royal Women’s Hospital, Parkville, Victoria, Australia;
; ORCID iD:
https://orcid.org/0000-0002-0132-9858
| |
Collapse
|
29
|
Aldehyde dehydrogenase 2-associated metabolic abnormalities and cardiovascular diseases: current status, underlying mechanisms, and clinical recommendations. CARDIOLOGY PLUS 2022. [DOI: 10.1097/cp9.0000000000000002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
30
|
Herr SA, Shi L, Gianaris T, Jiao Y, Sun S, Race N, Shapiro S, Shi R. Critical role of mitochondrial aldehyde dehydrogenase 2 in acrolein sequestering in rat spinal cord injury. Neural Regen Res 2021; 17:1505-1511. [PMID: 34916435 PMCID: PMC8771087 DOI: 10.4103/1673-5374.330613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lipid peroxidation-derived aldehydes, such as acrolein, the most reactive aldehyde, have emerged as key culprits in sustaining post-spinal cord injury (SCI) secondary pathologies leading to functional loss. Strong evidence suggests that mitochondrial aldehyde dehydrogenase-2 (ALDH2), a key oxidoreductase and powerful endogenous anti-aldehyde machinery, is likely important for protecting neurons from aldehydes-mediated degeneration. Using a rat model of spinal cord contusion injury and recently discovered ALDH2 activator (Alda-1), we planned to validate the aldehyde-clearing and neuroprotective role of ALDH2. Over an acute 2 day period post injury, we found that ALDH2 expression was significantly lowered post-SCI, but not so in rats given Alda-1. This lower enzymatic expression may be linked to heightened acrolein-ALDH2 adduction, which was revealed in co-immunoprecipitation experiments. We have also found that administration of Alda-1 to SCI rats significantly lowered acrolein in the spinal cord, and reduced cyst pathology. In addition, Alda-1 treatment also resulted in significant improvement of motor function and attenuated post-SCI mechanical hypersensitivity up to 28 days post-SCI. Finally, ALDH2 was found to play a critical role in in vitro protection of PC12 cells from acrolein exposure. It is expected that the outcome of this study will broaden and enhance anti-aldehyde strategies in combating post-SCI neurodegeneration and potentially bring treatment to millions of SCI victims. All animal work was approved by Purdue Animal Care and Use Committee (approval No. 1111000095) on January 1, 2021.
Collapse
Affiliation(s)
- Seth A Herr
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Liangqin Shi
- Department of Orthopedics, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Institute of Trauma and Orthopedics, Shanghai, China
| | - Thomas Gianaris
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yucheng Jiao
- Department of Orthopedics, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Institute of Trauma and Orthopedics, Shanghai, China
| | - Siyuan Sun
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Nick Race
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Scott Shapiro
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Riyi Shi
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
31
|
Calleja LF, Yoval-Sánchez B, Hernández-Esquivel L, Gallardo-Pérez JC, Sosa-Garrocho M, Marín-Hernández Á, Jasso-Chávez R, Macías-Silva M, Salud Rodríguez-Zavala J. Activation of ALDH1A1 by omeprazole reduces cell oxidative stress damage. FEBS J 2021; 288:4064-4080. [PMID: 33400378 DOI: 10.1111/febs.15698] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/30/2020] [Accepted: 01/04/2021] [Indexed: 11/30/2022]
Abstract
Under physiological conditions, cells produce low basal levels of reactive oxygen species (ROS); however, in pathologic conditions ROS production increases dramatically, generating high concentrations of toxic unsaturated aldehydes. Aldehyde dehydrogenases (ALDHs) are responsible for detoxification of these aldehydes protecting the cell. Due to the physiological relevance of these enzymes, it is important to design strategies to modulate their activity. It was previously reported that omeprazole activation of ALDH1A1 protected Escherichia coli cells overexpressing this enzyme, from oxidative stress generated by H2 O2 . In this work, omeprazole cell protection potential was evaluated in eukaryotic cells. AS-30D cell or hepatocyte suspensions were subjected to a treatment with omeprazole and exposure to light (that is required to activate omeprazole in the active site of ALDH) and then exposed to H2 O2 . Cells showed viability similar to control cells, total activity of ALDH was preserved, while cell levels of lipid aldehydes and oxidative stress markers were maintained low. Cell protection by omeprazole was avoided by inhibition of ALDHs with disulfiram, revealing the key role of these enzymes in the protection. Additionally, omeprazole also preserved ALDH2 (mitochondrial isoform) activity, diminishing lipid aldehyde levels and oxidative stress in this organelle, protecting mitochondrial respiration and transmembrane potential formation capacity, from the stress generated by H2 O2 . These results highlight the important role of ALDHs as part of the antioxidant system of the cell, since if the activity of these enzymes decreases under stress conditions, the viability of the cell is compromised.
Collapse
Affiliation(s)
- Luis Francisco Calleja
- Departamento de Bioquímica, Instituto Nacional de Cardiología 'Ignacio Chávez', Ciudad de México, México
| | - Belem Yoval-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología 'Ignacio Chávez', Ciudad de México, México
| | - Luz Hernández-Esquivel
- Departamento de Bioquímica, Instituto Nacional de Cardiología 'Ignacio Chávez', Ciudad de México, México
| | - Juan Carlos Gallardo-Pérez
- Departamento de Bioquímica, Instituto Nacional de Cardiología 'Ignacio Chávez', Ciudad de México, México
| | - Marcela Sosa-Garrocho
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Álvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología 'Ignacio Chávez', Ciudad de México, México
| | - Ricardo Jasso-Chávez
- Departamento de Bioquímica, Instituto Nacional de Cardiología 'Ignacio Chávez', Ciudad de México, México
| | - Marina Macías-Silva
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | | |
Collapse
|
32
|
The role of ALDH2 in tumorigenesis and tumor progression: Targeting ALDH2 as a potential cancer treatment. Acta Pharm Sin B 2021; 11:1400-1411. [PMID: 34221859 PMCID: PMC8245805 DOI: 10.1016/j.apsb.2021.02.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
A major mitochondrial enzyme for protecting cells from acetaldehyde toxicity is aldehyde dehydrogenase 2 (ALDH2). The correlation between ALDH2 dysfunction and tumorigenesis/growth/metastasis has been widely reported. Either low or high ALDH2 expression contributes to tumor progression and varies among different tumor types. Furthermore, the ALDH2∗2 polymorphism (rs671) is the most common single nucleotide polymorphism (SNP) in Asia. Epidemiological studies associate ALDH2∗2 with tumorigenesis and progression. This study summarizes the essential functions and potential ALDH2 mechanisms in the occurrence, progression, and treatment of tumors in various types of cancer. Our study indicates that ALDH2 is a potential therapeutic target for cancer therapy.
Collapse
Key Words
- 4-HNE, 4-hydroxy-2-nonenal
- ALD, alcoholic liver disease
- ALDH2
- ALDH2, aldehyde dehydrogenase 2
- AMPK, AMP-activated protein kinase
- Acetaldehyde
- BCa, bladder cancer
- COUP-TF, chicken ovalbumin upstream promoter-transcription factor
- CRC, colorectal cancer
- CSCs, cancer stem cells
- Cancer
- Cancer therapy
- DFS, disease-free survival
- EC, esophageal cancer
- FA, Fanconi anemia
- FANCD2, Fanconi anemia protein
- GCA, gastric cancer
- HCC, hepatocellular carcinoma
- HDACs, histone deacetylases
- HNC, head and neck cancer
- HNF-4, hepatocyte nuclear factor 4
- HR, homologous recombination
- LCSCs, liver cancer stem cells
- MDA, malondialdehyde
- MDR, multi-drug resistance
- MN, micronuclei
- Metastasis
- NAD, nicotinamide adenine dinucleotide
- NCEs, normochromic erythrocytes
- NER, nucleotide excision repair pathway
- NF-κB, nuclear factor-κB
- NHEJ, non-homologous end-joining
- NRF2, nuclear factor erythroid 2 (NF-E2)-related factor 2
- NRRE, nuclear receptor response element
- NSCLC, non-small-cell lung
- NeG, 1,N2-etheno-dGuo
- OPC, oropharyngeal cancer
- OS, overall survival
- OvCa, ovarian cancer
- PBMC, peripheral blood mononuclear cell
- PC, pancreatic cancer
- PdG, N2-propano-2′-deoxyguanosine
- Polymorphism
- Progression
- REV1, Y-family DNA polymerase
- SCC, squamous cell carcinoma
- TGF-β, transforming growth factor β
- Tumorigenesis
- VHL, von Hippel-Lindau
- ccRCC, clear-cell renal cell carcinomas
- εPKC, epsilon protein kinase C
Collapse
|
33
|
Wang Q, Chang B, Li X, Zou Z. Role of ALDH2 in Hepatic Disorders: Gene Polymorphism and Disease Pathogenesis. J Clin Transl Hepatol 2021; 9:90-98. [PMID: 33604259 PMCID: PMC7868706 DOI: 10.14218/jcth.2020.00104] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is a key enzyme of alcohol metabolism and it is involved in the cellular mechanism of alcohol liver disease. ALDH2 gene mutations exist in about 8% of the world's population, with the incidence reaching 45% in East Asia. The mutations will result in impairment of enzyme activity and accumulation of acetaldehyde, facilitating the progression of other liver diseases, including non-alcoholic fatty liver diseases, viral hepatitis and hepatocellular carcinoma, through adduct formation and inflammatory responses. In this review, we seek to summarize recent research progress on the correlation between ALDH2 gene polymorphism and multiple liver diseases, with an attempt to provide clues for better understanding of the disease mechanism and for strategy making.
Collapse
Affiliation(s)
- Qiaoling Wang
- Peking University, 302 Clinical Medical School, Beijing, China
- Diagnosis and Treatment Center for Non-Infectious Liver Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Binxia Chang
- Diagnosis and Treatment Center for Non-Infectious Liver Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoyan Li
- Anhui Medical University, Hefei, Anhui, China
| | - Zhengsheng Zou
- Peking University, 302 Clinical Medical School, Beijing, China
- Diagnosis and Treatment Center for Non-Infectious Liver Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Correspondence to: Zhengsheng Zou, The Center for Diagnosis and Treatment of Non-Infectious Liver Disease, The General Hospital of Chinese People’s Liberation Army No. 5 Medical Science Center, No. 100 Xisihuan Middle Road, Beijing 100039, China. E-mail:
| |
Collapse
|
34
|
Housh K, Jha JS, Haldar T, Amin SBM, Islam T, Wallace A, Gomina A, Guo X, Nel C, Wyatt JW, Gates KS. Formation and repair of unavoidable, endogenous interstrand cross-links in cellular DNA. DNA Repair (Amst) 2021; 98:103029. [PMID: 33385969 PMCID: PMC8882318 DOI: 10.1016/j.dnarep.2020.103029] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023]
Abstract
Genome integrity is essential for life and, as a result, DNA repair systems evolved to remove unavoidable DNA lesions from cellular DNA. Many forms of life possess the capacity to remove interstrand DNA cross-links (ICLs) from their genome but the identity of the naturally-occurring, endogenous substrates that drove the evolution and retention of these DNA repair systems across a wide range of life forms remains uncertain. In this review, we describe more than a dozen chemical processes by which endogenous ICLs plausibly can be introduced into cellular DNA. The majority involve DNA degradation processes that introduce aldehyde residues into the double helix or reactions of DNA with endogenous low molecular weight aldehyde metabolites. A smaller number of the cross-linking processes involve reactions of DNA radicals generated by oxidation.
Collapse
Affiliation(s)
- Kurt Housh
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Jay S Jha
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Tuhin Haldar
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Saosan Binth Md Amin
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Tanhaul Islam
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Amanda Wallace
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Anuoluwapo Gomina
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Xu Guo
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Christopher Nel
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Jesse W Wyatt
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States
| | - Kent S Gates
- University of Missouri, Department of Chemistry, 125 Chemistry Building, Columbia, MO 65211, United States; University of Missouri, Department of Biochemistry, Columbia, MO 65211, United States.
| |
Collapse
|
35
|
Oka Y, Hamada M, Nakazawa Y, Muramatsu H, Okuno Y, Higasa K, Shimada M, Takeshima H, Hanada K, Hirano T, Kawakita T, Sakaguchi H, Ichimura T, Ozono S, Yuge K, Watanabe Y, Kotani Y, Yamane M, Kasugai Y, Tanaka M, Suganami T, Nakada S, Mitsutake N, Hara Y, Kato K, Mizuno S, Miyake N, Kawai Y, Tokunaga K, Nagasaki M, Kito S, Isoyama K, Onodera M, Kaneko H, Matsumoto N, Matsuda F, Matsuo K, Takahashi Y, Mashimo T, Kojima S, Ogi T. Digenic mutations in ALDH2 and ADH5 impair formaldehyde clearance and cause a multisystem disorder, AMeD syndrome. SCIENCE ADVANCES 2020; 6:eabd7197. [PMID: 33355142 PMCID: PMC11206199 DOI: 10.1126/sciadv.abd7197] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
Rs671 in the aldehyde dehydrogenase 2 gene (ALDH2) is the cause of Asian alcohol flushing response after drinking. ALDH2 detoxifies endogenous aldehydes, which are the major source of DNA damage repaired by the Fanconi anemia pathway. Here, we show that the rs671 defective allele in combination with mutations in the alcohol dehydrogenase 5 gene, which encodes formaldehyde dehydrogenase (ADH5FDH ), causes a previously unidentified disorder, AMeD (aplastic anemia, mental retardation, and dwarfism) syndrome. Cellular studies revealed that a decrease in the formaldehyde tolerance underlies a loss of differentiation and proliferation capacity of hematopoietic stem cells. Moreover, Adh5-/-Aldh2 E506K/E506K double-deficient mice recapitulated key clinical features of AMeDS, showing short life span, dwarfism, and hematopoietic failure. Collectively, our results suggest that the combined deficiency of formaldehyde clearance mechanisms leads to the complex clinical features due to overload of formaldehyde-induced DNA damage, thereby saturation of DNA repair processes.
Collapse
Affiliation(s)
- Yasuyoshi Oka
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Motoharu Hamada
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuka Nakazawa
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Okuno
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koichiro Higasa
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mayuko Shimada
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Honoka Takeshima
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- School of Medicine, Nagoya University, Nagoya, Japan
| | - Katsuhiro Hanada
- Clinical Engineering Research Center, Faculty of Medicine, Oita University, Yufu, Japan
| | - Taichi Hirano
- Department of Hematology, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan
| | - Toshiro Kawakita
- Department of Hematology, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan
| | - Hirotoshi Sakaguchi
- Department of Hematology and Oncology, Children Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan
| | - Takuya Ichimura
- Department of Pediatrics, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Shuichi Ozono
- Department of Pediatrics and Child Health, School of Medicine, Kurume University, Kurume, Japan
| | - Kotaro Yuge
- Department of Pediatrics and Child Health, School of Medicine, Kurume University, Kurume, Japan
| | - Yoriko Watanabe
- Department of Pediatrics and Child Health, School of Medicine, Kurume University, Kurume, Japan
| | - Yuko Kotani
- Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
- Genome Editing Research and Development (R&D) Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mutsumi Yamane
- Center for Animal Research and Education, Nagoya University, Nagoya, Japan
| | - Yumiko Kasugai
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichiro Nakada
- Department of Bioregulation and Cellular Response, Graduate School of Medicine, Osaka University, Osaka, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan
| | - Norisato Mitsutake
- Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Yuichiro Hara
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kohji Kato
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiji Mizuno
- Department of Pediatrics, Aichi Developmental Disability Center, Kasugai, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yosuke Kawai
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Human Biosciences Unit for the Top Global Course Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Seiji Kito
- Center for Animal Research and Education, Nagoya University, Nagoya, Japan
| | - Keiichi Isoyama
- Department of Pediatrics, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Masafumi Onodera
- Division of Immunology, National Center for Child Health and Development, Tokyo, Japan
| | - Hideo Kaneko
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoji Mashimo
- Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
- Genome Editing Research and Development (R&D) Center, Graduate School of Medicine, Osaka University, Osaka, Japan
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiji Kojima
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan.
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
36
|
Acrolein: A Potential Mediator of Oxidative Damage in Diabetic Retinopathy. Biomolecules 2020; 10:biom10111579. [PMID: 33233661 PMCID: PMC7699716 DOI: 10.3390/biom10111579] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/05/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of vision loss among working-age adults. Extensive evidences have documented that oxidative stress mediates a critical role in the pathogenesis of DR. Acrolein, a product of polyamines oxidation and lipid peroxidation, has been demonstrated to be involved in the pathogenesis of various human diseases. Acrolein’s harmful effects are mediated through multiple mechanisms, including DNA damage, inflammation, ROS formation, protein adduction, membrane disruption, endoplasmic reticulum stress, and mitochondrial dysfunction. Recent investigations have reported the involvement of acrolein in the pathogenesis of DR. These studies have shown a detrimental effect of acrolein on the retinal neurovascular unit under diabetic conditions. The current review summarizes the existing literature on the sources of acrolein, the impact of acrolein in the generation of oxidative damage in the diabetic retina, and the mechanisms of acrolein action in the pathogenesis of DR. The possible therapeutic interventions such as the use of polyamine oxidase inhibitors, agents with antioxidant properties, and acrolein scavengers to reduce acrolein toxicity are also discussed.
Collapse
|
37
|
Yoval-Sánchez B, Calleja LF, de la Luz Hernández-Esquivel M, Rodríguez-Zavala JS. Piperlonguminine a new mitochondrial aldehyde dehydrogenase activator protects the heart from ischemia/reperfusion injury. Biochim Biophys Acta Gen Subj 2020; 1864:129684. [PMID: 32679250 DOI: 10.1016/j.bbagen.2020.129684] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Detoxification of aldehydes by aldehyde dehydrogenases (ALDHs) is crucial to maintain cell function. In cardiovascular diseases, reactive oxygen species generated during ischemia/reperfusion events trigger lipoperoxidation, promoting cell accumulation of highly toxic lipid aldehydes compromising cardiac function. In this context, activation of ALDH2, may contribute to preservation of cell integrity by diminishing aldehydes content more efficiently. METHODS The theoretic interaction of piperlonguminine (PPLG) with ALDH2 was evaluated by docking analysis. Recombinant human ALDH2 was used to evaluate the effects of PPLG on the kinetics of the enzyme. The effects of PPLG were further investigated in a myocardial infarction model in rats, evaluating ALDHs activity, antioxidant enzymes, oxidative stress markers and mitochondrial function. RESULTS PPLG increased the activity of recombinant human ALDH2 and protected the enzyme from inactivation by lipid aldehydes. Additionally, administration of this drug prevented the damage induced by ischemia/reperfusion in rats, restoring heart rate and blood pressure, which correlated with protection of ALDHs activity in the tissue, a lower content of lipid aldehydes, and the preservation of mitochondrial function. CONCLUSION Activation of ALDH2 by piperlonguminine ameliorates cell damage generated in heart ischemia/reperfusion events, by decreasing lipid aldehydes concentration promoting cardioprotection.
Collapse
Affiliation(s)
- Belem Yoval-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología "Ignacio Chávez", Ciudad de México, 14080, México
| | - Luis Francisco Calleja
- Departamento de Bioquímica, Instituto Nacional de Cardiología "Ignacio Chávez", Ciudad de México, 14080, México
| | | | - José Salud Rodríguez-Zavala
- Departamento de Bioquímica, Instituto Nacional de Cardiología "Ignacio Chávez", Ciudad de México, 14080, México.
| |
Collapse
|
38
|
Liu Z, Ye S, Zhong X, Wang W, Lai CH, Yang W, Yue P, Luo J, Huang X, Zhong Z, Xiong Y, Fan X, Li L, Wang Y, Ye Q. Pretreatment with the ALDH2 activator Alda‑1 protects rat livers from ischemia/reperfusion injury by inducing autophagy. Mol Med Rep 2020; 22:2373-2385. [PMID: 32705206 PMCID: PMC7411338 DOI: 10.3892/mmr.2020.11312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatic ischemia/reperfusion injury (HIRI) is a complex pathophysiological process that often leads to poor clinical prognosis. Clinically, the effective means to alleviate HIRI are limited. The aim of the present study was to investigate whether Alda-1, an activator of mitochondrial aldehyde dehydrogenase 2 (ALDH2), had a protective effect on HIRI and to investigate the mechanisms underlying this protective effect. Sprague-Dawley rats were treated with Alda-1 or Daidzin, an ALDH2 inhibitor, 30 min before partial (70%) warm liver ischemia to induce HIRI. The 48 rats were randomly divided into four groups: Sham, ischemia injury (IR), IR-Alda-1, and IR-Daidzin. After 6 and 24 h of reperfusion, serum and liver tissue samples were collected and stored for further experiments. Alanine aminotransferase, aspartate aminotransferase and hematoxylin & eosin staining was used to evaluate the liver damage. Western blotting and reverse transcription-quantitative PCR were used to detect the expression of related proteins and mRNA. TUNEL staining was used to observe the apoptosis of liver cells. Transmission electron microscopy was used to detect the mitochondrial injuries. Alda-1 pretreatment ameliorated the HIRI-induced damage to the liver function and reduced histological lesions. Alda-1 also increased ALDH2 activity after HIRI. Moreover, the pretreatment with Alda-1 reduced the accumulation of toxic aldehyde 4-hydroxy-2-nonenal, decreased the production of reactive oxygen species and malondialdehyde, reversed the damage to the liver mitochondria, attenuated hepatocyte apoptosis and inhibited the HIRI-induced inflammatory response, including high-mobility group box 1/toll-like receptor 4 signaling. Alda-1 also induced autophagy by upregulating autophagy-related 7 and Rab7 increasing the microtubule associated protein 1 light chain 3 αII/I ratio and inhibiting p62 expression. ALDH2-induced autophagy was dependent on the activation of the AKT/mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) signaling pathways. In conclusion, the findings of the present study suggested that Alda-1 may protect the liver against HIRI-induced damage, including hepatic enzyme injury, acetaldehyde accumulation, oxidative stress, hepatocyte apoptosis and inflammation. Alda-1 may confer this protection by inducing autophagy through the AKT/mTOR and AMPK signaling pathways. Therefore, ALDH2 could represent a potential pharmacological target in the clinical treatment of HIRI.
Collapse
Affiliation(s)
- Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Shaojun Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Xiang Zhong
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nangchang, Jiangxi 330006, P.R. China
| | - Wei Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Chin-Hui Lai
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Wang Yang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Pengpeng Yue
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Jun Luo
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Xiaoying Huang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Xiaoli Fan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Ling Li
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
39
|
Matsumura Y, Li N, Alwaseem H, Pagovich OE, Crystal RG, Greenblatt MB, Stiles KM. Systemic Adeno-Associated Virus-Mediated Gene Therapy Prevents the Multiorgan Disorders Associated with Aldehyde Dehydrogenase 2 Deficiency and Chronic Ethanol Ingestion. Hum Gene Ther 2020; 31:163-182. [PMID: 31801381 DOI: 10.1089/hum.2019.268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aldehyde dehydrogenase type 2 (ALDH2), a key enzyme in ethanol metabolism, processes toxic acetaldehyde to nontoxic acetate. ALDH2 deficiency affects 8% of the world population and 35-45% of East Asians. The ALDH2*2 allele common genetic variant has a glutamic acid-to-lysine substitution at position 487 (E487K) that reduces the oxidizing ability of the enzyme resulting in systemic accumulation of acetaldehyde with ethanol ingestion. With chronic ethanol ingestion, mutations in ALDH2 are associated with a variety of hematological, neurological, and dermatological abnormalities, and an increased risk for esophageal cancer and osteoporosis. Based on our prior studies demonstrating that a one-time administration of an adeno-associated virus (AAV) serotype rh.10 gene transfer vector expressing the human ALDH2 cDNA (AAVrh.10hALDH2) prevents the acute effects of ethanol administration (the "Asian flush syndrome"), we hypothesized that AAVrh.10hALDH2 would also prevent the chronic disorders associated with ALDH2 deficiency and chronic ethanol ingestion. To assess this hypothesis, AAVrh.10hALDH2 (1011 genome copies) was administered intravenously to two models of ALDH2 deficiency, Aldh2 knockout homozygous (Aldh2-/-) and knockin homozygous (Aldh2E487K+/+) mice (n = 10 per group). Four weeks after vector administration, mice were given drinking water with 10-15% ethanol for 12 weeks. Strikingly, compared with nonethanol drinking littermates, AAVrh.10hALDH2 administration prevented chronic ethanol-induced serum acetaldehyde accumulation and elevated liver malondialdehyde levels, loss of body weight, reduced hemoglobin levels, reduced performance in locomotor activity tests, accumulation of esophageal DNA damage and DNA adducts, and development of osteopenia. AAVrh.10hALDH2 should be considered as a preventative therapy for the increased risk of chronic disorders associated with ALDH2 deficiency and chronic alcohol exposure.
Collapse
Affiliation(s)
- Yuki Matsumura
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Na Li
- Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Hanan Alwaseem
- Proteomics Resource Center, The Rockefeller University, New York, New York
| | - Odelya E Pagovich
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Matthew B Greenblatt
- Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
- Research Division, Hospital for Special Surgery, New York, New York
| | - Katie M Stiles
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
40
|
Nannelli G, Ziche M, Donnini S, Morbidelli L. Endothelial Aldehyde Dehydrogenase 2 as a Target to Maintain Vascular Wellness and Function in Ageing. Biomedicines 2020; 8:E4. [PMID: 31947800 PMCID: PMC7168060 DOI: 10.3390/biomedicines8010004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/28/2019] [Accepted: 12/31/2019] [Indexed: 12/16/2022] Open
Abstract
Endothelial cells are the main determinants of vascular function, since their dysfunction in response to a series of cardiovascular risk factors is responsible for disease progression and further consequences. Endothelial dysfunction, if not resolved, further aggravates the oxidative status and vessel wall inflammation, thus igniting a vicious cycle. We have furthermore to consider the physiological manifestation of vascular dysfunction and chronic low-grade inflammation during ageing, also known as inflammageing. Based on these considerations, knowledge of the molecular mechanism(s) responsible for endothelial loss-of-function can be pivotal to identify novel targets of intervention with the aim of maintaining endothelial wellness and vessel trophism and function. In this review we have examined the role of the detoxifying enzyme aldehyde dehydrogenase 2 (ALDH2) in the maintenance of endothelial function. Its impairment indeed is associated with oxidative stress and ageing, and in the development of atherosclerosis and neurodegenerative diseases. Strategies to improve its expression and activity may be beneficial in these largely diffused disorders.
Collapse
Affiliation(s)
- Ginevra Nannelli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (G.N.); (S.D.)
| | - Marina Ziche
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (G.N.); (S.D.)
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (G.N.); (S.D.)
| |
Collapse
|
41
|
Chen N, Shan Q, Qi Y, Liu W, Tan X, Gu J. Transcriptome analysis in normal human liver cells exposed to 2, 3, 3', 4, 4', 5 - Hexachlorobiphenyl (PCB 156). CHEMOSPHERE 2020; 239:124747. [PMID: 31514003 DOI: 10.1016/j.chemosphere.2019.124747] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/31/2019] [Accepted: 09/03/2019] [Indexed: 05/20/2023]
Abstract
BACKGROUNDS Polychlorinated biphenyls are persistent environmental pollutants associated with the onset of non-alcoholic fatty liver disease in humans, but there is limited information on the underlying mechanism. In the present study, we investigated the alterations in gene expression profiles in normal human liver cells L-02 following exposure to 2, 3, 3', 4, 4', 5 - hexachlorobiphenyl (PCB 156), a potent compound that may induce non-alcoholic fatty liver disease. METHODS The L-02 cells were exposed to PCB 156 for 72 h and the contents of intracellular triacylglyceride and total cholesterol were subsequently measured. Microarray analysis of mRNAs and long non-coding RNAs (lncRNAs) in the cells was also performed after 3.4 μM PCB 156 treatment. RESULTS Exposure to PCB 156 (3.4 μM, 72 h) resulted in significant increases of triacylglyceride and total cholesterol concentrations in L-02 cells. Microarray analysis identified 222 differentially expressed mRNAs and 628 differentially expressed lncRNAs. Gene Ontology and pathway analyses associated the differentially expressed mRNAs with metabolic and inflammatory processes. Moreover, lncRNA-mRNA co-expression network revealed 36 network pairs comprising 10 differentially expressed mRNAs and 34 dysregulated lncRNAs. The results of bioinformatics analysis further indicated that dysregulated lncRNA NONHSAT174696, lncRNA NONHSAT179219, and lncRNA NONHSAT161887, as the regulators of EDAR, CYP1B1, and ALDH3A1 respectively, played an important role in the PCB 156-induced lipid metabolism disorder. CONCLUSION Our findings provide an overview of differentially expressed mRNAs and lncRNAs in L-02 cells exposed to PCB 156, and contribute to the field of polychlorinated biphenyl-induced non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Ningning Chen
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Qiuli Shan
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China; State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China.
| | - Yu Qi
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Wei Liu
- State Key Laboratory of Environmental Chemistry and Eco-Toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Xiaojun Tan
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| | - Jinsong Gu
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China
| |
Collapse
|
42
|
Koenders SA, Wijaya LS, Erkelens MN, Bakker AT, van der Noord VE, van Rooden EJ, Burggraaff L, Putter PC, Botter E, Wals K, van den Elst H, den Dulk H, Florea BI, van de Water B, van Westen GJP, Mebius RE, Overkleeft HS, Le Dévédec SE, van der Stelt M. Development of a Retinal-Based Probe for the Profiling of Retinaldehyde Dehydrogenases in Cancer Cells. ACS CENTRAL SCIENCE 2019; 5:1965-1974. [PMID: 31893226 PMCID: PMC6936097 DOI: 10.1021/acscentsci.9b01022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Indexed: 05/13/2023]
Abstract
Retinaldehyde dehydrogenases belong to a superfamily of enzymes that regulate cell differentiation and are responsible for detoxification of anticancer drugs. Chemical tools and methods are of great utility to visualize and quantify aldehyde dehydrogenase (ALDH) activity in health and disease. Here, we present the discovery of a first-in-class chemical probe based on retinal, the endogenous substrate of retinal ALDHs. We unveil the utility of this probe in quantitating ALDH isozyme activity in a panel of cancer cells via both fluorescence and chemical proteomic approaches. We demonstrate that our probe is superior to the widely used ALDEFLUOR assay to explain the ability of breast cancer (stem) cells to produce all-trans retinoic acid. Furthermore, our probe revealed the cellular selectivity profile of an advanced ALDH1A1 inhibitor, thereby prompting us to investigate the nature of its cytotoxicity. Our results showcase the application of substrate-based probes in interrogating pathologically relevant enzyme activities. They also highlight the general power of chemical proteomics in driving the discovery of new biological insights and its utility to guide drug discovery efforts.
Collapse
Affiliation(s)
- Sebastiaan
T. A. Koenders
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
- Oncode Institute, Utrecht 3521 AL, The Netherlands
| | - Lukas S. Wijaya
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Martje N. Erkelens
- Department
of Molecular Cell Biology and Immunology, Amsterdam University Medical Centra, Amsterdam 1081 HV, The Netherlands
| | - Alexander T. Bakker
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Vera E. van der Noord
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Eva J. van Rooden
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Lindsey Burggraaff
- Computational
Drug Discovery, Division of Drug Discovery and Safety, Leiden Academic
Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Pasquale C. Putter
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Else Botter
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Kim Wals
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
- Oncode Institute, Utrecht 3521 AL, The Netherlands
| | - Hans van den Elst
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Hans den Dulk
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Bogdan I. Florea
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Bob van de Water
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Gerard J. P. van Westen
- Computational
Drug Discovery, Division of Drug Discovery and Safety, Leiden Academic
Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Reina E. Mebius
- Department
of Molecular Cell Biology and Immunology, Amsterdam University Medical Centra, Amsterdam 1081 HV, The Netherlands
| | - Herman S. Overkleeft
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Sylvia E. Le Dévédec
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Mario van der Stelt
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
- Oncode Institute, Utrecht 3521 AL, The Netherlands
- E-mail:
| |
Collapse
|
43
|
Bozi LHM, Campos JC, Zambelli VO, Ferreira ND, Ferreira JCB. Mitochondrially-targeted treatment strategies. Mol Aspects Med 2019; 71:100836. [PMID: 31866004 DOI: 10.1016/j.mam.2019.100836] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
Disruption of mitochondrial function is a common feature of inherited mitochondrial diseases (mitochondriopathies) and many other infectious and non-infectious diseases including viral, bacterial and protozoan infections, inflammatory and chronic pain, neurodegeneration, diabetes, obesity and cardiovascular diseases. Mitochondria therefore become an attractive target for developing new therapies. In this review we describe critical mechanisms involved in the maintenance of mitochondrial functionality and discuss strategies used to identify and validate mitochondrial targets in different diseases. We also highlight the most recent preclinical and clinical findings using molecules targeting mitochondrial bioenergetics, morphology, number, content and detoxification systems in common pathologies.
Collapse
Affiliation(s)
- Luiz H M Bozi
- Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Juliane C Campos
- Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | | | | | - Julio C B Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Brazil; Department of Chemical and Systems Biology, School of Medicine, Stanford University, USA.
| |
Collapse
|
44
|
Matsumura Y, Stiles KM, Reid J, Frenk EZ, Cronin S, Pagovich OE, Crystal RG. Gene Therapy Correction of Aldehyde Dehydrogenase 2 Deficiency. Mol Ther Methods Clin Dev 2019; 15:72-82. [PMID: 31649957 PMCID: PMC6804850 DOI: 10.1016/j.omtm.2019.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) deficiency causes "Asian flush syndrome," presenting as alcohol-induced facial flushing, tachycardia, nausea, and headaches. One of the most common hereditary enzyme deficiencies, it affects 35%-40% of East Asians and 8% of the world population. ALDH2 is the key enzyme in ethanol metabolism; with ethanol challenge, the common ALDH2*2 (E487K) mutation results in accumulation of toxic acetaldehyde. ALDH2*2 heterozygotes have increased risk for upper digestive tract cancers, compounded by smoking and drinking alcohol. We hypothesized that a one-time administration of an adeno-associated virus (AAV) gene transfer vector expressing the human ALDH2 coding sequence (AAVrh.10hALDH2) would correct the deficiency state. AAVrh.10hALDH2 was administered intravenously to Aldh2 knockout (Aldh2 -/-) and Aldh2 E487K knockin homozygous (Aldh2 E487K+/+) mice. Following acute ethanol ingestion, untreated ALDH2-deficient mice had elevated acetaldehyde levels and performed poorly in behavioral tests. In contrast, treated Aldh2 -/- and Aldh2 E487K+/+ mice had lower serum acetaldehyde levels and improved behavior. Thus, in vivo AAV-mediated ALDH2 therapy may reverse the deficiency state in ALDH2*2 individuals, eliminating the Asian flush syndrome and reducing the risk for associated disorders.
Collapse
Affiliation(s)
- Yuki Matsumura
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Katie M. Stiles
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jasmine Reid
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Esther Z. Frenk
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Samantha Cronin
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Odelya E. Pagovich
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
45
|
Targeting liver aldehyde dehydrogenase-2 prevents heavy but not moderate alcohol drinking. Proc Natl Acad Sci U S A 2019; 116:25974-25981. [PMID: 31792171 DOI: 10.1073/pnas.1908137116] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aldehyde dehydrogenase 2 (ALDH2), a key enzyme for detoxification the ethanol metabolite acetaldehyde, is recognized as a promising therapeutic target to treat alcohol use disorders (AUDs). Disulfiram, a potent ALDH2 inhibitor, is an approved drug for the treatment of AUD but has clinical limitations due to its side effects. This study aims to elucidate the relative contribution of different organs in acetaldehyde clearance through ALDH2 by using global- (Aldh2 -/-) and tissue-specific Aldh2-deficient mice, and to examine whether liver-specific ALDH2 inhibition can prevent alcohol-seeking behavior. Aldh2 -/- mice showed markedly higher acetaldehyde concentrations than wild-type (WT) mice after acute ethanol gavage. Acetaldehyde levels in hepatocyte-specific Aldh2 knockout (Aldh2 Hep-/-) mice were significantly higher than those in WT mice post gavage, but did not reach the levels observed in Aldh2 -/- mice. Energy expenditure and motility were dramatically dampened in Aldh2 -/- mice, but moderately decreased in Aldh2 Hep-/- mice compared to controls. In the 2-bottle paradigm and the drinking-in-the-dark model, Aldh2 -/- mice drank negligible volumes from ethanol-containing bottles, whereas Aldh2 Hep-/- mice showed reduced alcohol preference at high but not low alcohol concentrations. Glial cell- or neuron-specific Aldh2 deficiency did not affect voluntary alcohol consumption. Finally, specific liver Aldh2 knockdown via injection of shAldh2 markedly decreased alcohol preference. In conclusion, although the liver is the major organ responsible for acetaldehyde metabolism, a cumulative effect of ALDH2 from other organs likely also contributes to systemic acetaldehyde clearance. Liver-targeted ALDH2 inhibition can decrease heavy drinking without affecting moderate drinking, providing molecular basis for hepatic ALDH2 targeting/editing for the treatment of AUD.
Collapse
|
46
|
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
|
47
|
Fonseca TL, Fernandes GW, Bocco BMLC, Keshavarzian A, Jakate S, Donohue TM, Gereben B, Bianco AC. Hepatic Inactivation of the Type 2 Deiodinase Confers Resistance to Alcoholic Liver Steatosis. Alcohol Clin Exp Res 2019; 43:1376-1383. [PMID: 30908637 PMCID: PMC6602874 DOI: 10.1111/acer.14027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/15/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND A mouse with hepatocyte-specific deiodinase type II inactivation (Alb-D2KO) is resistant to diet-induced obesity, hepatic steatosis, and hypertriglyceridemia due to perinatal epigenetic modifications in the liver. This phenotype is linked to low levels of Zfp125, a hepatic transcriptional repressor that promotes liver steatosis by inhibiting genes involved in packaging and secretion of very-low-density lipoprotein. METHODS Here, we used chronic and binge ethanol (EtOH) in mice to cause liver steatosis. RESULTS The EtOH treatment causes a 2.3-fold increase in hepatic triglyceride content; Zfp125 levels were approximately 50% higher in these animals. In contrast, Alb-D2KO mice did not develop EtOH-induced liver steatosis. They also failed to elevate Zfp125 to the same levels, despite being on the EtOH-containing diet for the same period of time. Their phenotype was associated with 1.3- to 2.9-fold up-regulation of hepatic genes involved in lipid transport and export that are normally repressed by Zfp125, that is, Mttp, Abca1, Ldlr, Apoc1, Apoc3, Apoe, Apoh, and Azgp1. Furthermore, genes involved in the EtOH metabolic pathway, that is, Aldh2 and Acss2, were also 1.6- to 3.1-fold up-regulated in Alb-D2KO EtOH mice compared with control animals kept on EtOH. CONCLUSIONS EtOH consumption elevates expression of Zfp125. Alb-D2KO animals, which have lower levels of Zfp125, are much less susceptible to EtOH-induced liver steatosis.
Collapse
Affiliation(s)
- Tatiana L. Fonseca
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL
| | - Gustavo W. Fernandes
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL
| | | | - Ali Keshavarzian
- Division of Digestive Diseases and Nutrition, Rush University, Chicago IL
| | | | - Terrence M. Donohue
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha NE
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Antonio C. Bianco
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL
| |
Collapse
|
48
|
Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol 2019; 23:101107. [PMID: 30692038 PMCID: PMC6859567 DOI: 10.1016/j.redox.2019.101107] [Citation(s) in RCA: 1230] [Impact Index Per Article: 205.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/13/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is a key regulator of the cellular antioxidant response, controlling the expression of genes that counteract oxidative and electrophilic stresses. Many pathological conditions are linked to imbalances in redox homeostasis, illustrating the important role of antioxidant defense systems in preventing the pathogenic effects associated with the accumulation of reactive species. In particular, it is becoming increasingly apparent that the accumulation of lipid peroxides has an important role in driving the pathogenesis of multiple disease states. A key example of this is the recent discovery of a novel form of cell death termed ferroptosis. Ferroptosis is an iron-dependent, lipid peroxidation-driven cell death cascade that has become a key target in the development of anti-cancer therapies, as well as the prevention of neurodegenerative and cardiovascular diseases. In this review, we will provide a brief overview of lipid peroxidation, as well as key components involved in the ferroptotic cascade. We will also highlight the role of the NRF2 signaling pathway in mediating lipid peroxidation and ferroptosis, focusing on established NRF2 target genes that mitigate these pathways, as well as the relevance of the NRF2-lipid peroxidation-ferroptosis axis in disease.
Collapse
Affiliation(s)
- Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Raul Castro-Portuguez
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721; Arizona Cancer Center, University of Arizona, Tucson, AZ, USA, 85724.
| |
Collapse
|
49
|
Chen L, Lang AL, Poff GD, Ding WX, Beier JI. Vinyl chloride-induced interaction of nonalcoholic and toxicant-associated steatohepatitis: Protection by the ALDH2 activator Alda-1. Redox Biol 2019; 24:101205. [PMID: 31026768 PMCID: PMC6479707 DOI: 10.1016/j.redox.2019.101205] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/10/2019] [Accepted: 04/14/2019] [Indexed: 12/20/2022] Open
Abstract
Vinyl chloride (VC), an abundant environmental contaminant causes steatohepatitis at high levels, but is considered safe at lower (i.e., sub-OSHA) levels. However, we have previously shown that even lower VC levels exacerbate experimental nonalcoholic fatty liver disease (NAFLD) caused by high-fat diet (HFD). Mitochondrial oxidative injury and subsequent metabolic dysfunction appeared to play key roles in mediating this interaction. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) serves as a key line of defense against endogenous and exogenous reactive aldehydes. The current study therefore tests the hypothesis that allosteric activation of ALDH2 with Alda-1 will protect against VC-enhanced NAFLD. Mice were exposed to low VC concentrations (<1 ppm), or room air for 6 h/day, 5 days/week for 12 weeks, while on HFD or low-fat control diet (LFD). Some mice received Alda-1 (20 mg/kg i.p., 3 × /week) for the last 3 weeks of diet/VC exposure. Indices of liver injury, oxidative stress, metabolic and mitochondrial (dys)function were measured. As observed previously, low-dose VC did not cause liver injury in control mice; while liver injury caused by HFD was enhanced by VC. VC decreased hepatic ALDH2 activity of mice fed HFD. Alda-1 attenuated oxidative stress, liver injury, and dysmetabolism in mice exposed to HFD+VC under these conditions. Importantly, alterations in mitochondrial function caused by VC and HFD were diminished by Alda-1. Previous studies have indicated that liver injury caused by HFD is mediated, at least in part, by enhanced mitochondrial autophagy (mitophagy). Here, Alda-1 suppressed PINK1/PARKIN-mediated mitophagy. Taken together, these results support the hypothesis that ALDH2 is a critical defense against mitochondrial injury caused by VC in experimental NAFLD. The ALDH2 activator Alda-1 conferred protection against liver damage under these conditions, most likely via increasing clearance of aldehydes and preserving mitochondrial respiratory function. VC, combined with HFD impairs ALDH2 function, causing an accumulation of endogenous aldehydes and oxidative stress in vivo. VC metabolite chloroacetaldehyde directly blocks ALDH2 activity in vitro. Alda-1 treatment reverses pre-established liver injury, oxidative stress and metabolic dysregulation caused by VC and HFD. Alda-1 increases overall autophagy caused by VC+HFD, but decreases mitophagy, likely to preserve mitochondrial function.
Collapse
Affiliation(s)
- Liya Chen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA; Hepatobiology and Toxicology Program, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA; University of Louisville Alcohol Research Center, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA.
| | - Anna L Lang
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA; Hepatobiology and Toxicology Program, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA; University of Louisville Alcohol Research Center, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA.
| | - Gavin D Poff
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA; Hepatobiology and Toxicology Program, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA; University of Louisville Alcohol Research Center, University of Louisville Health Sciences Center, Louisville, KY, 40202, USA.
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Juliane I Beier
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
50
|
Adeniji EA, Olotu FA, Shunmugam L, Soliman MES. From a computational point of view: deciphering the molecular synergism between oxidative stress-induced lipid peroxidation products and metabolic dysfunctionality of human liver mitochondrial aldehyde dehydrogenase-2. MOLECULAR SIMULATION 2019. [DOI: 10.1080/08927022.2019.1578355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Emmanuel A. Adeniji
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Fisayo A. Olotu
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Letitia Shunmugam
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mahmoud E. S. Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|