1
|
Kwok HC, Tse HT, Ng KK, Wang S, Au CK, Cai Z, Chan W. Absorptivity Is an Important Determinant in the Toxicity Difference between Aristolochic Acid I and Aristolochic Acid II. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2551-2561. [PMID: 39808478 PMCID: PMC11800392 DOI: 10.1021/acs.jafc.4c10765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Inadvertent exposure to aristolochic acids (AAs) is causing chronic renal disease worldwide, with aristolochic acid I (AA-I) identified as the primary toxic agent. This study employed chemical methods to investigate the mechanisms underlying the nephrotoxicity and carcinogenicity of AA-I. Aristolochic acid II (AA-II), which has a structure similar to that of AA-I, was investigated with the same methods for comparison. Despite their structural similarities, findings from cultured human cells and gut sac experiments showed that AA-I is absorbed more effectively than AA-II (∼3 times greater for AA-I than for AA-II; p < 0.001). This increased absorption, along with the previously observed higher activity of reductive activation enzymes for AA-I, results in greater DNA damage and oxidative stress, both of which are key factors in AA-related toxicity. The similar patterns of cell mortality (34.4 ± 2.3% vs 9.7 ± 0.1% for AA-I and AA-II at 80 μM; p < 0.0001), DNA adduct formation (∼3 times greater for AA-I than for AA-II; p < 0.001), and oxidative stress levels in relation to the concentrations of AA-I and AA-II indicate that the higher absorption rate of AA-I is a significant contributor to its greater toxicity. The toxicity of AA-I was also found to be further enhanced by its (natural) coexistence with AA-II. Since AA-I and AA-II differ only by a methoxy group, future research on reducing risks associated with AA exposure should focus on strategies to lower the absorption of these compounds.
Collapse
Affiliation(s)
- Hong-Ching Kwok
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Hei-Tak Tse
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Ka-Ki Ng
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Shuangshuang Wang
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Chun-Kit Au
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Zongwei Cai
- Eastern
Institute of Technology Ningbo, Ningbo, Zhejiang 315200, China
- Department
of Chemistry and State Key Laboratory of Environmental and Biological
Analysis, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
| | - Wan Chan
- Department
of Chemistry, The Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
2
|
Biyabani A, Ghorbani F, Koushki M, Nedaei K, Hemmati M, Mahdei Nasir Mahalleh N, Ghadimi D. Quercetin and calorie restriction improve leptin/adiponectin balance through reducing high-fat diet-induced oxidative stress in male BALB/c mice. Biochem Biophys Res Commun 2025; 742:151073. [PMID: 39637705 DOI: 10.1016/j.bbrc.2024.151073] [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: 08/13/2024] [Revised: 10/29/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Throughout the recent decades, obesity has become a serious health problem that raises the risk of several diseases, including cancer, diabetes, hypertension, heart disease, neurological musculoskeletal disorders, and Non-alcoholic fatty liver disease. Some strategies, such as dietary interventions, calorie restriction (CR), and the use of antioxidant compounds, have been proposed to improve quality of life in relation to obesity. The goal of this study was to characterize the effects of CR and quercetin (QUER) on obesity-induced oxidative stress (OS). Thirty 8-week-old male BALB/c mice were divided into 5 groups of six mice each: normal diet, high-fat diet (HFD), HFD + CR, HFD + QUER (15 mg kg-1, IP), and HFD + QUER + CR. CR was applied as two fasting days with an interval of two days in a week. Catalase (CAT), Paraxonase 1 (PON1) and adiponectin (APN) were decreased in the HFD group, while the combination of QUER and CR increased these parameters. Treatment with QUER and CR improved Alanine transaminase and Alkaline Phosphatase enzyme activity and also the amount of leptin and insulin. Moreover, combined QUER and CR also reduced triacylglycerol (TAG), total cholesterol and TAG droplets in hepatocytes. Decreased OS was associated with the higher expression of NAD(P)H Quinone Oxidoreductase 1(NQO1) and reduced hepatic vacuoles in QUER and CR-HFD treated groups. In conclusion, these findings suggest that the combination of QUER and CR might exert protective impacts on obesity through alleviating OS and the regulation of metabolism.
Collapse
Affiliation(s)
- Arezou Biyabani
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fereshte Ghorbani
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Koushki
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Keivan Nedaei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mina Hemmati
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Nima Mahdei Nasir Mahalleh
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Darya Ghadimi
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
3
|
Alhaji JH, Pathak D, Ashfaq F, Alsayegh AA, Khatoon F, Almutairi BJ, Khan MI, Beg MMA. Role of NQO1 Gene Involvement and Susceptibility of T2DM Among Saudi Arabia Population. Rejuvenation Res 2024; 27:145-153. [PMID: 38959119 DOI: 10.1089/rej.2024.0032] [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] [Indexed: 07/05/2024] Open
Abstract
NQO1 disruption enhances susceptibility to oxidative stress during hyperglycemia and is a significant contributor to the development and progression of diabetes. Oxidative stress has been linked to several symptoms, including hyperglycemia, reactive oxygen species buildup, high blood pressure, and the expression of inflammatory markers. Therefore, the present research aimed to evaluate the genetic abnormality of NQO1 (rs1800566, C609T) gene polymorphism, expression, and vitamin-D level assessment among Type 2 diabetes mellitus (T2DM) patients. The study included 100 newly diagnosed T2DM cases and 100 healthy individuals as healthy controls. Total RNA was extracted from the whole blood using the TRIzol method, and further cDNA was synthesized, and expression was evaluated. There is a significant difference in NQO1 (rs1800566, C609T) genotype distribution among the T2DM patients and healthy controls (p = 0.04). Compared with the NQO1 CC wild-type genotype, the NQO1 CT heterozygous genotype had an odds ratio of 1.96 (1.08-3.55), and the NQO1 TT mutant type genotype had an odds ratio of 3.31 (0.61-17.77). Significantly decreased expression of NQO1 mRNA was observed with heterozygous CT (p < 0.0001) and homozygous mutant TT genotype (p = 0.0004), compared with homozygous wild-type CC genotype. NQO1 mRNA expression level was also compared with vitamin D levels among the T2DM patients. T2DM patients with vitamin D deficiency had 1.83-fold NQO1 mRNA expression, while vitamin D insufficient and sufficient T2DM cases had 3.31-fold (p < 0.0001) and 3.70-fold (p < 0.0001) NQO1 mRNA expression. It was concluded that NQO1 (rs1800566, C609T) CT and TT genotypes played a significant role in the worseness of type II diabetes mellitus, and decreased expression of NQO1 mRNA expression could be an essential factor for disease worseness as well as hypermethylation could be a factor for reduced expression leading to disease severity. The decreased NQO1 mRNA expression with heterozygous CT and mutant TT genotype associated with vitamin D deficiency may contribute to disease progression.
Collapse
Affiliation(s)
- Jwaher Haji Alhaji
- Department of Health Sciences, College of Applied Studies and Community Service, King Saud University, Riyadh, Saudi Arabia
| | - Divya Pathak
- Central Drugs Standard Control Organisation, New Delhi, India
| | - Fauzia Ashfaq
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan, Saudi Arabia
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan, Saudi Arabia
| | - Fahmida Khatoon
- Department of Biochemistry, College of Medicine, University of Ha'il, Ha'il, Saudi Arabia
| | | | - Mohammad Idreesh Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mirza Masroor Ali Beg
- Faculty of Medicine, Alatoo International University, Bishkek, Kyrgyzstan
- Center for Promotion of Medical Research, Alatoo International University, Bishkek, Kyrgyzstan
| |
Collapse
|
4
|
Grieco A, Quereda-Moraleda I, Martin-Garcia JM. Innovative Strategies in X-ray Crystallography for Exploring Structural Dynamics and Reaction Mechanisms in Metabolic Disorders. J Pers Med 2024; 14:909. [PMID: 39338163 PMCID: PMC11432794 DOI: 10.3390/jpm14090909] [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: 07/22/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024] Open
Abstract
Enzymes are crucial in metabolic processes, and their dysfunction can lead to severe metabolic disorders. Structural biology, particularly X-ray crystallography, has advanced our understanding of these diseases by providing 3D structures of pathological enzymes. However, traditional X-ray crystallography faces limitations, such as difficulties in obtaining suitable protein crystals and studying protein dynamics. X-ray free-electron lasers (XFELs) have revolutionized this field with their bright and brief X-ray pulses, providing high-resolution structures of radiation-sensitive and hard-to-crystallize proteins. XFELs also enable the study of protein dynamics through room temperature structures and time-resolved serial femtosecond crystallography, offering comprehensive insights into the molecular mechanisms of metabolic diseases. Understanding these dynamics is vital for developing effective therapies. This review highlights the contributions of protein dynamics studies using XFELs and synchrotrons to metabolic disorder research and their application in designing better therapies. It also discusses G protein-coupled receptors (GPCRs), which, though not enzymes, play key roles in regulating physiological systems and are implicated in many metabolic disorders.
Collapse
Affiliation(s)
| | | | - Jose Manuel Martin-Garcia
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry Blas Cabrera, Spanish National Research Council (CSIC), 28006 Madrid, Spain; (A.G.); (I.Q.-M.)
| |
Collapse
|
5
|
Xiao JL, Liu HY, Sun CC, Tang CF. Regulation of Keap1-Nrf2 signaling in health and diseases. Mol Biol Rep 2024; 51:809. [PMID: 39001962 DOI: 10.1007/s11033-024-09771-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) functions as a central regulator in modulating the activities of diverse antioxidant enzymes, maintaining cellular redox balance, and responding to oxidative stress (OS). Kelch-like ECH-associated protein 1 (Keap1) serves as a principal negative modulator in controlling the expression of detoxification and antioxidant genes. It is widely accepted that OS plays a pivotal role in the pathogenesis of various diseases. When OS occurs, leading to inflammatory infiltration of neutrophils, increased secretion of proteases, and the generation of large quantities of reactive oxygen radicals (ROS). These ROS can oxidize or disrupt DNA, lipids, and proteins either directly or indirectly. They also cause gene mutations, lipid peroxidation, and protein denaturation, all of which can result in disease. The Keap1-Nrf2 signaling pathway regulates the balance between oxidants and antioxidants in vivo, maintains the stability of the intracellular environment, and promotes cell growth and repair. However, the antioxidant properties of the Keap1-Nrf2 signaling pathway are reduced in disease. This review overviews the mechanisms of OS generation, the biological properties of Keap1-Nrf2, and the regulatory role of its pathway in health and disease, to explore therapeutic strategies for the Keap1-Nrf2 signaling pathway in different diseases.
Collapse
Affiliation(s)
- Jiang-Ling Xiao
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan, 410012, China
| | - Heng-Yuan Liu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan, 410012, China
| | - Chen-Chen Sun
- Institute of Physical Education, Hunan First Normal University, Changsha, Hunan, 410205, China.
| | - Chang-Fa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan, 410012, China.
| |
Collapse
|
6
|
Mondal S, Basu S, Ghosh S, Guria S, Mukherjee S. Diethyl phthalate, a plasticizer, induces adipocyte inflammation and apoptosis in mice after long-term dietary administration. J Biochem Mol Toxicol 2024; 38:e23561. [PMID: 37942807 DOI: 10.1002/jbt.23561] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 09/06/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023]
Abstract
The incidence of metabolic diseases is increasing alarmingly in recent times. Parallel to nutritional excess and sedentary lifestyle, the random usage of several endocrine disrupting chemicals including plasticizers is reported to be closely associated with metabolic diseases. Diethyl phthalate (DEP) is a widely used plasticizer in a host of consumer and daily care products. Adipose tissue plays a central role in energy storage and whole-body metabolism. The impairment of adipose function is critically implicated in the pathogenesis of insulin resistance, diabetes, and related metabolic diseases. Recently, exposure to certain phthalate esters has been linked to the development of obesity and diabetes, although there are contradictions and the mechanisms are not clearly understood. In an effort to ascertain the metabolic consequences of chronic phthalate exposure and the underlying mechanism, the present study was designed to examine the effects of long-term dietary consumption of DEP in adipocytes. DEP-treated mice were hyperglycemic but nonobese; their body weight initially increased which subsequently was reduced compared to control. DEP exposure at lower levels impaired adipogenesis by downregulating the key transcription factor, peroxisome proliferator-activated receptor γ and its downstream insulin-sensitizing adipokine, adiponectin, thereby severely compromising adipocyte function. The activation of master regulator nuclear factor κB led to rise in proinflammatory cytokines. We found that DEP triggered intrinsic apoptotic pathways through activated cytochrome c-Apaf1-caspase 9-caspase 3 axis in adipocytes. Taken together, our data revealed that chronic administration of dietary DEP could unleash adverse metabolic outcomes by initiating oxidative stress, inflammation, and apoptosis in the adipocytes, thus leading to adipose tissue dysfunction.
Collapse
Affiliation(s)
- Shirsha Mondal
- Department of Zoology, Endocrinology and Metabolism Laboratory, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
| | - Soumyadeep Basu
- Department of Zoology, Endocrinology and Metabolism Laboratory, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
| | - Songita Ghosh
- Department of Zoology, Endocrinology and Metabolism Laboratory, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
| | - Suktara Guria
- Department of Zoology, Endocrinology and Metabolism Laboratory, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
| | - Sutapa Mukherjee
- Department of Zoology, Endocrinology and Metabolism Laboratory, Visva-Bharati (A Central University), Santiniketan, West Bengal, India
| |
Collapse
|
7
|
Abulmeaty MMA, Ghneim HK, Almajwal AM, Razak S, AlSahli AS, Andrade Laborde JE, Ibrahim EM, Aboul-Soud MAM, Al-Sheikh YA. Effects of unpredictable chronic mild stress on the cellular redox state and mitochondrial energy homeostasis in rat adipose tissue: A comprehensive metabolic study. Cell Biochem Funct 2023; 41:1330-1342. [PMID: 37805950 DOI: 10.1002/cbf.3867] [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: 06/27/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
Unpredictable chronic mild stress (UCMS) leads to variable metabolic effects. Oxidative stress (OS) of adipose tissue (AT) and mitochondrial energy homeostasis is little investigated. This work studied the effects of UCMS on OS and the antioxidant/redox status in AT and mitochondrial energy homeostasis in rats. Twenty-four male Wistar rats (180-220 g) were divided into two equal groups; the normal control (NC) group and the UCMS group which were exposed to various stresses for 28 days. An indirect calorimetry machine was used to measure volumes of respiratory gases (VO2 & VCO2 ), total energy expenditure (TEE), and food intake (FI). The AT depots were collected, weighed, and used for measuring activities and gene expression of key antioxidant enzymes (GPx1, SOD, CAT, GR, GCL, and GS), OS marker levels including superoxide anion (SA), peroxynitrite radical (PON), nitric oxide (NO), hydrogen peroxide (H2 O2 ), lipid peroxides (LPO), t-protein carbonyl content (PCC), and reduced/oxidized glutathione levels (GSH, GSSG). Additionally, AT mitochondrial fractions were used to determine the activities of the tricarboxylic acid cycle (TCA) cycle enzymes (CS, α-KGDH, ICDH, SDH, MDH), respiratory chain complexes I-III, II-III, IV, the nicotinamide coenzymes NAD+ , NADH, and ATP/ADP levels. Compared with the NC group, the UCMS group showed very significantly increased OS marker levels, lowered antioxidant enzyme activities and gene expression, as well as lowered TCA cycle and respiratory chain activity and NAD+ , NADH, and ATP levels (p < .001 for all comparisons). Besides, the UCMS group had lowered TEE and insignificant FI and weight gain. In conclusion, AT of the UCMS-subjected rats showed a state of disturbed redox balance linked to disrupted energy homeostasis producing augmentation of AT.
Collapse
Affiliation(s)
- Mahmoud M A Abulmeaty
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia
| | - Hazem K Ghneim
- Chair of Medical and Molecular Genetics, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia
| | - Ali Madi Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia
| | - Abdulaziz S AlSahli
- Chair of Medical and Molecular Genetics, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia
| | - Juan E Andrade Laborde
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida, USA
| | - Ebtesam M Ibrahim
- Department of Medical Physiology, School of Medicine, Zagazig University, Zagazig, Egypt
| | - Mourad A M Aboul-Soud
- Chair of Medical and Molecular Genetics, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia
| | - Yazeed A Al-Sheikh
- Chair of Medical and Molecular Genetics, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia
| |
Collapse
|
8
|
Xu F, Ziebarth JD, Goeminne LJ, Gao J, Williams EG, Quarles LD, Makowski L, Cui Y, Williams RW, Auwerx J, Lu L. Gene network based analysis identifies a coexpression module involved in regulating plasma lipids with high-fat diet response. J Nutr Biochem 2023; 119:109398. [PMID: 37302664 PMCID: PMC10896179 DOI: 10.1016/j.jnutbio.2023.109398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/08/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Plasma lipids are modulated by gene variants and many environmental factors, including diet-associated weight gain. However, understanding how these factors jointly interact to influence molecular networks that regulate plasma lipid levels is limited. Here, we took advantage of the BXD recombinant inbred family of mice to query weight gain as an environmental stressor on plasma lipids. Coexpression networks were examined in both nonobese and obese livers, and a network was identified that specifically responded to the obesogenic diet. This obesity-associated module was significantly associated with plasma lipid levels and enriched with genes known to have functions related to inflammation and lipid homeostasis. We identified key drivers of the module, including Cidec, Cidea, Pparg, Cd36, and Apoa4. The Pparg emerged as a potential master regulator of the module as it can directly target 19 of the top 30 hub genes. Importantly, activation of this module is causally linked to lipid metabolism in humans, as illustrated by correlation analysis and inverse-variance weighed Mendelian randomization. Our findings provide novel insights into gene-by-environment interactions for plasma lipid metabolism that may ultimately contribute to new biomarkers, better diagnostics, and improved approaches to prevent or treat dyslipidemia in patients.
Collapse
Affiliation(s)
- Fuyi Xu
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China; Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jesse D Ziebarth
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ludger Je Goeminne
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Lausanne, Switzerland
| | - Jun Gao
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Evan G Williams
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Leigh D Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Liza Makowski
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yan Cui
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Robert W Williams
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Lausanne, Switzerland.
| | - Lu Lu
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| |
Collapse
|
9
|
Wakabayashi N, Yagishita Y, Joshi T, Kensler TW. Forced Hepatic Expression of NRF2 or NQO1 Impedes Hepatocyte Lipid Accumulation in a Lipodystrophy Mouse Model. Int J Mol Sci 2023; 24:13345. [PMID: 37686150 PMCID: PMC10487640 DOI: 10.3390/ijms241713345] [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: 07/09/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Lipodystrophy is a disorder featuring loss of normal adipose tissue depots due to impaired production of normal adipocytes. It leads to a gain of fat deposition in ectopic tissues such as liver and skeletal muscle that results in steatosis, dyslipidemia, and insulin resistance. Previously, we established a Rosa NIC/NIC::AdiCre lipodystrophy model mouse. The lipodystrophic phenotype that included hepatomegaly accompanied with hepatic damage due to higher lipid accumulation was attenuated substantially by amplified systemic NRF2 signaling in mice with hypomorphic expression of Keap1; whole-body Nrf2 deletion abrogated this protection. To determine whether hepatic-specific NRF2 signaling would be sufficient for protection against hepatomegaly and fatty liver development, direct, powerful, transient expression of Nrf2 or its target gene Nqo1 was achieved by administration through hydrodynamic tail vein injection of pCAG expression vectors of dominant-active Nrf2 and Nqo1 in Rosa NIC/NIC::AdiCre mice fed a 9% fat diet. Both vectors enabled protection from hepatic damage, with the pCAG-Nqo1 vector being the more effective as seen with a ~50% decrease in hepatic triglyceride levels. Therefore, activating NRF2 signaling or direct elevation of NQO1 in the liver provides new possibilities to partially reduce steatosis that accompanies lipodystrophy.
Collapse
Affiliation(s)
- Nobunao Wakabayashi
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA or (Y.Y.); (T.J.); (T.W.K.)
| | - Yoko Yagishita
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA or (Y.Y.); (T.J.); (T.W.K.)
- Division of Endocrinology, Columbia University, New York, NY 10032, USA
| | - Tanvi Joshi
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA or (Y.Y.); (T.J.); (T.W.K.)
| | - Thomas W. Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA or (Y.Y.); (T.J.); (T.W.K.)
| |
Collapse
|
10
|
Qiu D, Song S, Chen N, Bian Y, Yuan C, Zhang W, Duan H, Shi Y. NQO1 alleviates renal fibrosis by inhibiting the TLR4/NF-κB and TGF-β/Smad signaling pathways in diabetic nephropathy. Cell Signal 2023; 108:110712. [PMID: 37196773 DOI: 10.1016/j.cellsig.2023.110712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/19/2023]
Abstract
OBJECTIVE Diabetic nephropathy (DN) is one of the main complications of diabetes, and inflammation and fibrosis play an important role in its progression. NAD(P)H: quinone oxidoreductase 1 (NQO1) protects cells from oxidative stress and damage caused by toxic quinones. In the present study, we aimed to investigate the protective effects of NQO1 against diabetes-induced renal inflammation and fibrosis and the underlying mechanisms. METHODS In vivo, the kidneys of type 2 diabetes model db/db mice were infected with adeno-associated virus vectors to induce NQO1 overexpression. In vitro, human renal tubular epithelial (HK-2) cells transfected with NQO1 pcDNA3.1(+) were cultured under high-glucose (HG) conditions. Gene and protein expression was assessed by quantitative real-time PCR, Western blotting, immunofluorescence, and immunohistochemical staining. Mitochondrial reactive oxygen species (ROS) were detected with MitoSOX Red. RESULT Our study revealed that the expression of NQO1 was markedly downregulated and that Toll-like receptor (TLR)4 and TGF-β1 expression was upregulated in vivo and in vitro under diabetic conditions. Overexpression of NQO1 suppressed proinflammatory cytokine (IL-6, TNF-α, MCP-1) secretion, extracellular matrix (ECM) (collagen IV, fibronectin) accumulation and epithelial-mesenchymal transition (EMT) (α-SMA, E-cadherin) in the db/db mouse kidneys and HG-cultured HK-2 cells. Furthermore, NQO1 overexpression ameliorated HG-induced TLR4/NF-κB and TGF-β/Smad pathways activation. Mechanistic studies demonstrated that a TLR4 inhibitor (TAK-242) suppressed the TLR4/NF-κB signaling pathway, proinflammatory cytokine secretion, EMT and ECM-related protein expression in HG-exposed HK-2 cells. In addition, we found that the antioxidants N-acetylcysteine (NAC) and tempol increased the expression of NQO1 and decreased the expression of TLR4, TGF-β1, Nox1, and Nox4 and ROS production in HK-2 cells cultured under HG conditions. CONCLUSIONS These data suggest that NQO1 alleviates diabetes-induced renal inflammation and fibrosis by regulating the TLR4/NF-κB and TGF-β/Smad signaling pathways.
Collapse
Affiliation(s)
- Duojun Qiu
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Department of Pathology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Ning Chen
- Department of Pathology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yawei Bian
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Chen Yuan
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Wei Zhang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Huijun Duan
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China.
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China.
| |
Collapse
|
11
|
Salama AAA, Elgohary R, Fahmy MI. Protocatechuic acid ameliorates lipopolysaccharide-induced kidney damage in mice via downregulation of TLR-4-mediated IKBKB/NF-κB and MAPK/Erk signaling pathways. J Appl Toxicol 2023. [PMID: 36807594 DOI: 10.1002/jat.4447] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Acute kidney injury (AKI) is a very critical cause of death in the whole world. Lipopolysaccharide (LPS) induces kidney damage by activating various deleterious inflammatory and oxidative pathways. Protocatechuic acid, a natural phenolic compound, has shown to exert beneficial effects against oxidative and inflammatory responses. The study aimed to clarify the nephroprotective activity of protocatechuic acid in LPS-induced acute kidney damage in mice. Forty male Swiss mice were allocated in four groups as follows: normal control group; LPS (250 μg/kg, ip)-induced kidney injury group; LPS-injected mice treated with protocatechuic acid (15 mg/kg, po), and LPS-injected mice treated with protocatechuic acid (30 mg/kg, po). Significant toll-like receptor 4 (TLR-4)-mediated activation of IKBKB/NF-κB and MAPK/Erk/COX-2 inflammatory pathways has been observed in kidneys of mice treated with LPS. Oxidative stress was revealed by inhibition of total antioxidant capacity, catalase, nuclear factor erythroid 2-related factor 2 (Nrf2), and NAD(P)H quinone oxidoreductase (NQO1) enzyme along with increased nitric oxide level. In parallel, focal inflammatory effects were shown in between the tubules and glomeruli as well as in the perivascular dilated blood vessels at the cortex affecting the normal morphology of the kidney tissues of LPS-treated mice. However, treatment with protocatechuic acid reduced LPS-induced changes in the aforementioned parameters and restored normal histological features of the affected tissues. In conclusion, our study uncovered that protocatechuic acid has nephroprotective effects in mice with AKI through opposing different inflammatory and oxidative cascades.
Collapse
Affiliation(s)
| | - Rania Elgohary
- Narcotics, Ergogenics and Poisons Department, National Research Centre, Cairo, Egypt
| | - Mohamed Ibrahim Fahmy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
12
|
Han B, Kikuta S, Kamogashira T, Kondo K, Yamasoba T. Sleep deprivation induces delayed regeneration of olfactory sensory neurons following injury. Front Neurosci 2022; 16:1029279. [DOI: 10.3389/fnins.2022.1029279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
The circadian system, which is essential for the alignment of sleep/wake cycles, modulates adult neurogenesis. The olfactory epithelium (OE) has the ability to generate new neurons throughout life. Loss of olfactory sensory neurons (OSNs) as a result of injury to the OE triggers the generation of new OSNs, which are incorporated into olfactory circuits to restore olfactory sensory perception. This regenerative potential means that it is likely that the OE is substantially affected by sleep deprivation (SD), although how this may occur remains unclear. The aim of this study is to address how SD affects the process of OSN regeneration following OE injury. Mice were subjected to SD for 2 weeks, which induced changes in circadian activity. This condition resulted in decreased activity during the night-time and increased activity during the daytime, and induced no histological changes in the OE. However, when subjected to SD during the regeneration process after OE injury, a significant decrease in the number of mature OSNs in the dorsomedial area of the OE, which is the only area containing neurons expressing NQO1 (quinone dehydrogenase 1), was observed compared to the NQO1-negative OE. Furthermore, a significant decrease in proliferating basal cells was observed in the NQO1-positive OE compared to the NQO1-negative OE, but no increase in apoptotic OSNs was observed. These results indicate that SD accompanied by disturbed circadian activity could induce structurally negative effects on OSN regeneration, preferentially in the dorsomedial area of the OE, and that this area-specific regeneration delay might involve the biological activity of NQO1.
Collapse
|
13
|
Antioxidant properties of 3-hydroxy-2-ethyl-6-methylpyridinium nitroxysuccinate upon the activation of oxidative processes by antitumor drug Cisplatin in vitro and in vivo. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3692-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
14
|
Sibiya T, Ghazi T, Mohan J, Nagiah S, Chuturgoon AA. Spirulina platensis Ameliorates Oxidative Stress Associated with Antiretroviral Drugs in HepG2 Cells. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11223143. [PMID: 36432871 PMCID: PMC9694780 DOI: 10.3390/plants11223143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 06/01/2023]
Abstract
Lately, Spirulina platensis (SP), as an antioxidant, has exhibited high potency in the treatment of oxidative stress, diabetes, immune disorder, inflammatory stress, and bacterial and viral-related diseases. This study investigated the possible protective role of Spirulina platensis against ARV-induced oxidative stress in HepG2 cells. Human liver (HepG2) cells were treated with ARVs ((Lamivudine (3TC): 1.51 µg/mL, tenofovir disoproxil fumarate (TDF): 0.3 µg/mL and Emtricitabine (FTC): 1.8 µg/mL)) for 96 h and thereafter treated with 1.5 µg/mL Spirulina platensis for 24 h. After the treatments, the gene and protein expressions of the antioxidant response pathway were determined using a quantitative polymerase chain reaction (qPCR) and Western blots. The results show that Spirulina platensis decreased the gene expressions of Akt (p < 0.0001) and eNOS (↓p < 0.0001) while, on the contrary, it increased the transcript levels of NRF-2 (↑p = 0.0021), Keap1 (↑p = 0.0002), CAT (↑p < 0.0001), and NQO-1 (↑p = 0.1432) in the HepG2 cells. Furthermore, the results show that Spirulina platensis also decreased the protein expressions of NRF-2 (↓p = 0.1226) and pNRF-2 (↓p = 0.0203). Interestingly, HAART-SP induced an NRF-2 pathway response through upregulating NRF-2 (except for FTC-SP) (↑p < 0.0001), CAT (↑p < 0.0001), and NQO-1 (except for FTC-SP) (↑p < 0.0001) mRNA expression. In addition, NRF-2 (↑p = 0.0085) and pNRF-2 (↑p < 0.0001) protein expression was upregulated in the HepG2 cells post-exposure to HAART-SP. The results, therefore, allude to the fact that Spirulina platensis has the potential to mitigate HAART-adverse drug reactions (HAART toxicity) through the activation of antioxidant response in HepG2 cells. We hereby recommend further studies on Spirulina platensis and HAART synergy.
Collapse
Affiliation(s)
- Thabani Sibiya
- Discipline of Medical Biochemistry and Chemical Pathology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Terisha Ghazi
- Discipline of Medical Biochemistry and Chemical Pathology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Jivanka Mohan
- Discipline of Medical Biochemistry and Chemical Pathology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Savania Nagiah
- Discipline of Medical Biochemistry and Chemical Pathology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4013, South Africa
- Medical Programme, Department of Human Biology, Faculty of Health Sciences, Nelson Mandela University Missionvale, Port Elizabeth 6059, South Africa
| | - Anil A. Chuturgoon
- Discipline of Medical Biochemistry and Chemical Pathology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Howard College Campus, University of KwaZulu-Natal, Durban 4013, South Africa
| |
Collapse
|
15
|
Poljšak B, Kovač V, Milisav I. Current Uncertainties and Future Challenges Regarding NAD+ Boosting Strategies. Antioxidants (Basel) 2022; 11:1637. [PMID: 36139711 PMCID: PMC9495723 DOI: 10.3390/antiox11091637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Precursors of nicotinamide adenine dinucleotide (NAD+), modulators of enzymes of the NAD+ biosynthesis pathways and inhibitors of NAD+ consuming enzymes, are the main boosters of NAD+. Increasing public awareness and interest in anti-ageing strategies and health-promoting lifestyles have grown the interest in the use of NAD+ boosters as dietary supplements, both in scientific circles and among the general population. Here, we discuss the current trends in NAD+ precursor usage as well as the uncertainties in dosage, timing, safety, and side effects. There are many unknowns regarding pharmacokinetics and pharmacodynamics, particularly bioavailability, metabolism, and tissue specificity of NAD+ boosters. Given the lack of long-term safety studies, there is a need for more clinical trials to determine the proper dose of NAD+ boosters and treatment duration for aging prevention and as disease therapy. Further research will also need to address the long-term consequences of increased NAD+ and the best approaches and combinations to increase NAD+ levels. The answers to the above questions will contribute to the more efficient and safer use of NAD+ boosters.
Collapse
Affiliation(s)
- Borut Poljšak
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Vito Kovač
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Irina Milisav
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
16
|
Kanayama M, Luo J. CD38-Induced Apoptosis and Mitochondrial Damage is Restored by Nicotinamide in Prostate Cancer. Front Mol Biosci 2022; 9:890402. [PMID: 35677882 PMCID: PMC9168365 DOI: 10.3389/fmolb.2022.890402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/03/2022] [Indexed: 11/25/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential molecule for living organisms. CD38 is a key NAD+-dependent enzyme which breaks down NAD+ to cyclic ADP-ribose (ADPR) and nicotinamide (NAM, vitamin B3), and NAM can be recycled to synthesize NAD+. CD38 expression is consistently silenced by methylation in prostate cancer and progressively downregulated in advanced castration-resistant prostate cancer, suggesting a connection between NAD+ and prostate carcinogenesis as well as prostate cancer progression. However, the functional interplay between NAD+, CD38, and NAM remains largely uncharacterized in prostate cancer cells. In this study, we generated stable LNCaP95 cell clones expressing varying levels of CD38 upon induction by doxycycline. We demonstrate that CD38 overexpression resulted in growth suppression and apoptosis accompanied by cleavage of poly (ADP-ribose) polymerase 1 (PARP1). CD38 overexpression also dramatically reduced intracellular NAD+ levels and decreased mitochondrial respiration as measured by oxygen consumption rate. We further show that some but not all of these CD38-induced phenotypes could be rescued by exogenous NAM. Treatment of cells with NAM rescued CD38-induced apoptosis and mitochondrial stress but did not restore intracellular NAD+ levels. We also found that NAM demonstrated biphasic effect on mitochondria function, a finding that can be explained by the dual role of NAM as both a precursor of NAD+ and also as a suppressor of a number of NAD+-dependent enzymes. Collectively, these findings provide additional insight supporting the functional relevance of CD38 loss in prostate cancer by linking cell-autonomous regulation of mitochondrial function and prostate cancer.
Collapse
|
17
|
Rodriguez-Iturbe B, Johnson RJ, Lanaspa MA, Nakagawa T, Garcia-Arroyo FE, Sánchez-Lozada LG. Sirtuin deficiency and the adverse effects of fructose and uric acid synthesis. Am J Physiol Regul Integr Comp Physiol 2022; 322:R347-R359. [PMID: 35271385 PMCID: PMC8993531 DOI: 10.1152/ajpregu.00238.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 12/17/2022]
Abstract
Fructose metabolism and hyperuricemia have been shown to drive insulin resistance, metabolic syndrome, hepatic steatosis, hypertension, inflammation, and innate immune reactivity in experimental studies. We suggest that these adverse effects are at least in part the result of suppressed activity of sirtuins, particularly Sirtuin1. Deficiency of sirtuin deacetylations is a consequence of reduced bioavailability of its cofactor nicotinamide adenine dinucleotide (NAD+). Uric acid-induced inflammation and oxidative stress consume NAD+ and activation of the polyol pathway of fructose and uric acid synthesis also reduces the NAD+-to-NADH ratio. Variability in the compensatory regeneration of NAD+ could result in variable recovery of sirtuin activity that may explain the inconsistent benefits of treatments directed to reduce uric acid in clinical trials. Here, we review the pathogenesis of the metabolic dysregulation driven by hyperuricemia and their potential relationship with sirtuin deficiency. In addition, we discuss therapeutic options directed to increase NAD+ and sirtuins activity that may improve the adverse effects resulting from fructose and uric acid synthesis.
Collapse
Affiliation(s)
- Bernardo Rodriguez-Iturbe
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán," Mexico City, Mexico
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Denver, Colorado
- Kidney Disease Division, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, Colorado
| | - Miguel A Lanaspa
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, Oregon
| | | | - Fernando E Garcia-Arroyo
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| | - Laura G Sánchez-Lozada
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| |
Collapse
|
18
|
Qiu D, Song S, Wang Y, Bian Y, Wu M, Wu H, Shi Y, Duan H. NAD(P)H: quinone oxidoreductase 1 attenuates oxidative stress and apoptosis by regulating Sirt1 in diabetic nephropathy. J Transl Med 2022; 20:44. [PMID: 35090502 PMCID: PMC8796493 DOI: 10.1186/s12967-021-03197-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/14/2021] [Indexed: 12/29/2022] Open
Abstract
Background Diabetic nephropathy (DN) is one of the main complications of diabetes, and oxidative stress plays an important role in its progression. NAD(P)H: quinone oxidoreductase 1 (NQO1) protects cells from oxidative stress and toxic quinone damage. In the present study, we aimed to investigate the protective effects and underlying mechanisms of NQO1 on diabetes-induced renal tubular epithelial cell oxidative stress and apoptosis. Methods In vivo, the kidneys of db/db mice, which are a type 2 diabetes model, were infected with adeno-associated virus to induce NQO1 overexpression. In vitro, human renal tubular epithelial cells (HK-2 cells) were transfected with NQO1 pcDNA3.1(+) and cultured in high glucose (HG). Gene and protein expression was assessed by quantitative real-time PCR, western blotting, immunofluorescence analysis, and immunohistochemical staining. Reactive oxygen species (ROS) were examined by MitoSox red and flow cytometry. TUNEL assays were used to measure apoptosis. Result In vivo, NQO1 overexpression reduced the urinary albumin/creatinine ratio (UACR) and blood urea nitrogen (BUN) level in db/db mice. Our results revealed that NQO1 overexpression could significantly increase the ratio of NAD+/NADH and silencing information regulator 1 (Sirt1) expression and block tubular oxidative stress and apoptosis in diabetic kidneys. In vitro, NQO1 overexpression reduced the generation of ROS, NADPH oxidase 1 (Nox1) and Nox4, the Bax/Bcl-2 ratio and the expression of Cleaved Caspase-3 and increased NAD+/NADH levels and Sirt1 expression in HK-2 cells under HG conditions. However, these effects were reversed by the Sirt1 inhibitor EX527. Conclusions All these data suggest that NQO1 has a protective effect against oxidative stress and apoptosis in DN, which may be mediated by the regulation of Sirt1 through increasing intracellular NAD+/NADH levels. Therefore, NQO1 may be a new therapeutic target for DN.
Collapse
Affiliation(s)
- Duojun Qiu
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China
| | - Yuhan Wang
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China.,Digestive Department, Tangshan Workers Hospital, Tangshan, China
| | - Yawei Bian
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China
| | - Ming Wu
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China
| | - Haijiang Wu
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China. .,Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China.
| | - Huijun Duan
- Department of Pathology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, China. .,Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China.
| |
Collapse
|
19
|
Levine DC, Kuo HY, Hong HK, Cedernaes J, Hepler C, Wright AG, Sommars MA, Kobayashi Y, Marcheva B, Gao P, Ilkayeva OR, Omura C, Ramsey KM, Newgard CB, Barish GD, Peek CB, Chandel NS, Mrksich M, Bass J. NADH inhibition of SIRT1 links energy state to transcription during time-restricted feeding. Nat Metab 2021; 3:1621-1632. [PMID: 34903884 PMCID: PMC8688143 DOI: 10.1038/s42255-021-00498-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/28/2021] [Indexed: 11/08/2022]
Abstract
In mammals, circadian rhythms are entrained to the light cycle and drive daily oscillations in levels of NAD+, a cosubstrate of the class III histone deacetylase sirtuin 1 (SIRT1) that associates with clock transcription factors. Although NAD+ also participates in redox reactions, the extent to which NAD(H) couples nutrient state with circadian transcriptional cycles remains unknown. Here we show that nocturnal animals subjected to time-restricted feeding of a calorie-restricted diet (TRF-CR) only during night-time display reduced body temperature and elevated hepatic NADH during daytime. Genetic uncoupling of nutrient state from NADH redox state through transduction of the water-forming NADH oxidase from Lactobacillus brevis (LbNOX) increases daytime body temperature and blood and liver acyl-carnitines. LbNOX expression in TRF-CR mice induces oxidative gene networks controlled by brain and muscle Arnt-like protein 1 (BMAL1) and peroxisome proliferator-activated receptor alpha (PPARα) and suppresses amino acid catabolic pathways. Enzymatic analyses reveal that NADH inhibits SIRT1 in vitro, corresponding with reduced deacetylation of SIRT1 substrates during TRF-CR in vivo. Remarkably, Sirt1 liver nullizygous animals subjected to TRF-CR display persistent hypothermia even when NADH is oxidized by LbNOX. Our findings reveal that the hepatic NADH cycle links nutrient state to whole-body energetics through the rhythmic regulation of SIRT1.
Collapse
Affiliation(s)
- Daniel C Levine
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hsin-Yu Kuo
- Departments of Chemistry, Biomedical Engineering, and Cell and Molecular Biology, Northwestern University, Evanston, IL, USA
| | - Hee-Kyung Hong
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jonathan Cedernaes
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Departments of Medical Sciences and Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Chelsea Hepler
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alexandra G Wright
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Meredith A Sommars
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yumiko Kobayashi
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Biliana Marcheva
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, USA
| | - Chiaki Omura
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kathryn M Ramsey
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christopher B Newgard
- Duke Molecular Physiology Institute, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, USA
| | - Grant D Barish
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Clara Bien Peek
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Milan Mrksich
- Departments of Chemistry, Biomedical Engineering, and Cell and Molecular Biology, Northwestern University, Evanston, IL, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
20
|
Lee WS, Ham W, Kim J. Roles of NAD(P)H:quinone Oxidoreductase 1 in Diverse Diseases. Life (Basel) 2021; 11:life11121301. [PMID: 34947831 PMCID: PMC8703842 DOI: 10.3390/life11121301] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 01/07/2023] Open
Abstract
NAD(P)H:quinone oxidoreductase (NQO) is an antioxidant flavoprotein that catalyzes the reduction of highly reactive quinone metabolites by employing NAD(P)H as an electron donor. There are two NQO enzymes—NQO1 and NQO2—in mammalian systems. In particular, NQO1 exerts many biological activities, including antioxidant activities, anti-inflammatory effects, and interactions with tumor suppressors. Moreover, several recent studies have revealed the promising roles of NQO1 in protecting against cardiovascular damage and related diseases, such as dyslipidemia, atherosclerosis, insulin resistance, and metabolic syndrome. In this review, we discuss recent developments in the molecular regulation and biochemical properties of NQO1, and describe the potential beneficial roles of NQO1 in diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
- Correspondence: (W.-S.L.); (J.K.); Tel.: +82-2-6299-1419 (W.-S.L.); +82-2-6299-1397 (J.K.)
| | - Woojin Ham
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
- Correspondence: (W.-S.L.); (J.K.); Tel.: +82-2-6299-1419 (W.-S.L.); +82-2-6299-1397 (J.K.)
| |
Collapse
|
21
|
Basu P, Kim JH, Saeed S, Martins-Green M. Using systems biology approaches to identify signalling pathways activated during chronic wound initiation. Wound Repair Regen 2021; 29:881-898. [PMID: 34536049 DOI: 10.1111/wrr.12963] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/18/2021] [Accepted: 08/04/2021] [Indexed: 12/30/2022]
Abstract
Chronic wounds are a significant health problem worldwide. However, nothing is known about how chronic wounds initiate and develop. Here we use a chronic wound model in diabetic mice and a Systems Biology Approach using nanoString nCounter technology and weighted gene correlation network analysis (WGCNA), with tissues collected at 6, 12, 24 and 48 h post-wounding, to identify metabolic signalling pathways involved in initiation of chronicity. Normalized counts obtained from the nanoString nCounter Mouse Metabolic Panel were used for the WGCNA, which groups genes into co-expression modules to visualize the correlation network. Genes with significant module membership and gene trait significance (p < 0.05) were used to identify signalling pathways that are important for the development of chronicity. The pathway analysis using the Reactome database showed stabilization of PTEN, which down-regulates PI3K/AKT1, which in turn down-regulates Nrf2, as shown by ELISA, thus disabling antioxidant production, resulting in high oxidative stress levels. We find that pathways involved in inflammation, including those that generate pro-inflammatory lipids derived from arachidonic acid metabolism, IFNγ and catecholamines, occur. Moreover, HIF3α is over-expressed, potentially blocking Hif1α and preventing activation of growth factors and cytokines that promote granulation tissue formation. We also find that FGF1 is under-expressed, while thrombospondin-1 is over-expressed, resulting in decreased angiogenesis, a process that is critical for healing. Finally, enzymes involved in glycolysis are down-regulated, resulting in decreased production of pyruvate, a molecule critical for ATP production, leading to extensive cell death and wound paralysis. These findings offer new avenues of study that may lead to the development of novel treatments of CW to be administered right after debridement.
Collapse
Affiliation(s)
- Proma Basu
- Department of Molecular, Cell and Systems Biology, UC, Riverside, California, USA
| | - Jane Hannah Kim
- Department of Molecular, Cell and Systems Biology, UC, Riverside, California, USA
| | - Shayan Saeed
- Department of Molecular, Cell and Systems Biology, UC, Riverside, California, USA
| | | |
Collapse
|
22
|
Li G, Liu H, Feng R, Kang TS, Wang W, Ko CN, Wong CY, Ye M, Ma DL, Wan JB, Leung CH. A bioactive ligand-conjugated iridium(III) metal-based complex as a Keap1-Nrf2 protein-protein interaction inhibitor against acetaminophen-induced acute liver injury. Redox Biol 2021; 48:102129. [PMID: 34526248 PMCID: PMC8710994 DOI: 10.1016/j.redox.2021.102129] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/30/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
Hepatotoxicity caused by an overdose of acetaminophen (APAP) is the leading reason for acute drug-related liver failure. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a protein that helps to regulate redox homeostasis and coordinate stress responses via binding to the Kelch-like ECH-associated protein 1 (Keap1). Targeting the Keap1-Nrf2 interaction has recently emerged as a potential strategy to alleviate liver injury caused by APAP. Here, we designed and synthesized a number of iridium (III) and rhodium (III) complexes bearing ligands with reported activity against oxidative stress, which is associated with Nrf2 transcriptional activation. The iridium (III) complex 1 bearing a bioactive ligand 2,9-dimethyl-1,10-phenanthroline and 4-chloro-2-phenylquinoline, a derivative of the bioactive ligand 2-phenylquinoline, was identified as a direct small-molecule inhibitor of the Keap1–Nrf2 protein-protein interaction. 1 could stabilize Keap1 protein, upregulate HO-1 and NQO1, and promote Nrf2 nuclear translocation in normal liver cells. Moreover, 1 reversed APAP-induced liver damage by disrupting Keap1–Nrf2 interaction and without inducing organ damage and immunotoxicity in mice. Our study demonstrates the identification of a selective and efficacious antagonist of Keap1–Nrf2 interaction possessed good cellular permeability in cellulo and ideal pharmacokinetic parameters in vivo, and, more importantly, validates the feasibility of conjugating metal complexes with bioactive ligands to generate metal-based drug leads as non-toxic Keap1–Nrf2 interaction inhibitors for treating APAP-induced acute liver injury. 1 reversed APAP-induced liver damage by disrupting Keap1–Nrf2 interaction without inducing organ damage or immunotoxicity. Complex 1 possessed good cellular permeability in cellulo and ideal pharmacokinetic parameters in vivo. Conjugating metal complexes with bioactive ligands opens a novel avenue for the treatment of APAP-induced liver damage.
Collapse
Affiliation(s)
- Guodong Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Hao Liu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Ruibing Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Tian-Shu Kang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Wanhe Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China; Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Chung-Nga Ko
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Chun-Yuen Wong
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong SAR, China
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China.
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
23
|
Tsvetkov P, Adler J, Strobelt R, Adamovich Y, Asher G, Reuven N, Shaul Y. NQO1 Binds and Supports SIRT1 Function. Front Pharmacol 2021; 12:671929. [PMID: 34234670 PMCID: PMC8255383 DOI: 10.3389/fphar.2021.671929] [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] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/19/2021] [Indexed: 11/13/2022] Open
Abstract
Silent information regulator 2-related enzyme 1 (SIRT1) is an NAD+-dependent class III deacetylase and a key component of the cellular metabolic sensing pathway. The requirement of NAD+ for SIRT1 activity led us to assume that NQO1, an NADH oxidoreductase producing NAD+, regulates SIRT1 activity. We show here that SIRT1 is capable of increasing NQO1 (NAD(P)H Dehydrogenase Quinone 1) transcription and protein levels. NQO1 physically interacts with SIRT1 but not with an enzymatically dead SIRT1 H363Y mutant. The interaction of NQO1 with SIRT1 is markedly increased under mitochondrial inhibition. Interestingly, under this condition the nuclear pool of NQO1 is elevated. Depletion of NQO1 compromises the role of SIRT1 in inducing transcription of several target genes and eliminates the protective role of SIRT1 following mitochondrial inhibition. Our results suggest that SIRT1 and NQO1 form a regulatory loop where SIRT1 regulates NQO1 expression and NQO1 binds and mediates the protective role of SIRT1 during mitochondrial stress. The interplay between an NADH oxidoreductase enzyme and an NAD+ dependent deacetylase may act as a rheostat in sensing mitochondrial stress.
Collapse
Affiliation(s)
- Peter Tsvetkov
- Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Julia Adler
- Department of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| | - Romano Strobelt
- Department of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| | - Yaarit Adamovich
- Department of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| | - Gad Asher
- Department of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| | - Nina Reuven
- Department of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Shaul
- Department of Molecular Genetics Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
24
|
Ghanem MS, Monacelli F, Nencioni A. Advances in NAD-Lowering Agents for Cancer Treatment. Nutrients 2021; 13:1665. [PMID: 34068917 PMCID: PMC8156468 DOI: 10.3390/nu13051665] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential redox cofactor, but it also acts as a substrate for NAD-consuming enzymes, regulating cellular events such as DNA repair and gene expression. Since such processes are fundamental to support cancer cell survival and proliferation, sustained NAD production is a hallmark of many types of neoplasms. Depleting intratumor NAD levels, mainly through interference with the NAD-biosynthetic machinery, has emerged as a promising anti-cancer strategy. NAD can be generated from tryptophan or nicotinic acid. In addition, the "salvage pathway" of NAD production, which uses nicotinamide, a byproduct of NAD degradation, as a substrate, is also widely active in mammalian cells and appears to be highly exploited by a subset of human cancers. In fact, research has mainly focused on inhibiting the key enzyme of the latter NAD production route, nicotinamide phosphoribosyltransferase (NAMPT), leading to the identification of numerous inhibitors, including FK866 and CHS-828. Unfortunately, the clinical activity of these agents proved limited, suggesting that the approaches for targeting NAD production in tumors need to be refined. In this contribution, we highlight the recent advancements in this field, including an overview of the NAD-lowering compounds that have been reported so far and the related in vitro and in vivo studies. We also describe the key NAD-producing pathways and their regulation in cancer cells. Finally, we summarize the approaches that have been explored to optimize the therapeutic response to NAMPT inhibitors in cancer.
Collapse
Affiliation(s)
- Moustafa S. Ghanem
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (M.S.G.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
25
|
Ross D, Siegel D. The diverse functionality of NQO1 and its roles in redox control. Redox Biol 2021; 41:101950. [PMID: 33774477 PMCID: PMC8027776 DOI: 10.1016/j.redox.2021.101950] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022] Open
Abstract
In this review, we summarize the multiple functions of NQO1, its established roles in redox processes and potential roles in redox control that are currently emerging. NQO1 has attracted interest due to its roles in cell defense and marked inducibility during cellular stress. Exogenous substrates for NQO1 include many xenobiotic quinones. Since NQO1 is highly expressed in many solid tumors, including via upregulation of Nrf2, the design of compounds activated by NQO1 and NQO1-targeted drug delivery have been active areas of research. Endogenous substrates have also been proposed and of relevance to redox stress are ubiquinone and vitamin E quinone, components of the plasma membrane redox system. Established roles for NQO1 include a superoxide reductase activity, NAD+ generation, interaction with proteins and their stabilization against proteasomal degradation, binding and regulation of mRNA translation and binding to microtubules including the mitotic spindles. We also summarize potential roles for NQO1 in regulation of glucose and insulin metabolism with relevance to diabetes and the metabolic syndrome, in Alzheimer's disease and in aging. The conformation and molecular interactions of NQO1 can be modulated by changes in the pyridine nucleotide redox balance suggesting that NQO1 may function as a redox-dependent molecular switch.
Collapse
Affiliation(s)
- David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
26
|
Role of PGC-1α in the Mitochondrial NAD + Pool in Metabolic Diseases. Int J Mol Sci 2021; 22:ijms22094558. [PMID: 33925372 PMCID: PMC8123861 DOI: 10.3390/ijms22094558] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play vital roles, including ATP generation, regulation of cellular metabolism, and cell survival. Mitochondria contain the majority of cellular nicotinamide adenine dinucleotide (NAD+), which an essential cofactor that regulates metabolic function. A decrease in both mitochondria biogenesis and NAD+ is a characteristic of metabolic diseases, and peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) orchestrates mitochondrial biogenesis and is involved in mitochondrial NAD+ pool. Here we discuss how PGC-1α is involved in the NAD+ synthesis pathway and metabolism, as well as the strategy for increasing the NAD+ pool in the metabolic disease state.
Collapse
|
27
|
Di Francesco A, Choi Y, Bernier M, Zhang Y, Diaz-Ruiz A, Aon MA, Kalafut K, Ehrlich MR, Murt K, Ali A, Pearson KJ, Levan S, Preston JD, Martin-Montalvo A, Martindale JL, Abdelmohsen K, Michel CR, Willmes DM, Henke C, Navas P, Villalba JM, Siegel D, Gorospe M, Fritz K, Biswal S, Ross D, de Cabo R. NQO1 protects obese mice through improvements in glucose and lipid metabolism. NPJ Aging Mech Dis 2020; 6:13. [PMID: 33298924 PMCID: PMC7678866 DOI: 10.1038/s41514-020-00051-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic nutrient excess leads to metabolic disorders and insulin resistance. Activation of stress-responsive pathways via Nrf2 activation contributes to energy metabolism regulation. Here, inducible activation of Nrf2 in mice and transgenesis of the Nrf2 target, NQO1, conferred protection from diet-induced metabolic defects through preservation of glucose homeostasis, insulin sensitivity, and lipid handling with improved physiological outcomes. NQO1-RNA interaction mediated the association with and inhibition of the translational machinery in skeletal muscle of NQO1 transgenic mice. NQO1-Tg mice on high-fat diet had lower adipose tissue macrophages and enhanced expression of lipogenic enzymes coincident with reduction in circulating and hepatic lipids. Metabolomics data revealed a systemic metabolic signature of improved glucose handling, cellular redox, and NAD+ metabolism while label-free quantitative mass spectrometry in skeletal muscle uncovered a distinct diet- and genotype-dependent acetylation pattern of SIRT3 targets across the core of intermediary metabolism. Thus, under nutritional excess, NQO1 transgenesis preserves healthful benefits.
Collapse
Affiliation(s)
- Andrea Di Francesco
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Calico Life Sciences, South San Francisco, CA, USA
| | - Youngshim Choi
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Yingchun Zhang
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, 475004, People's Republic of China
| | - Alberto Diaz-Ruiz
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Nutritional Interventions Group, Precision Nutrition and Aging, Institute IMDEA Food, Crta. de Canto Blanco n° 8, 28049, Madrid, Spain
| | - Miguel A Aon
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Krystle Kalafut
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Margaux R Ehrlich
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Department Food Science, Cornell University, Ithaca, NY, 14850, USA
| | - Kelsey Murt
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Ahmed Ali
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kevin J Pearson
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Sophie Levan
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Joshua D Preston
- Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
- Emory University School of Medicine (MD/PhD program), Atlanta, GA, USA
| | - Alejandro Martin-Montalvo
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Cole R Michel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Diana M Willmes
- Molecular Diabetology, Paul Langerhans Institute Dresden of the Helmholtz German Center for Diabetes Research Munich, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, 01307, Dresden, Germany
| | - Christine Henke
- Molecular Diabetology, Paul Langerhans Institute Dresden of the Helmholtz German Center for Diabetes Research Munich, University Hospital Carl Gustav Carus and Faculty of Medicine, TU Dresden, 01307, Dresden, Germany
| | - Placido Navas
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, 41013, Sevilla, Spain
| | - Jose Manuel Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Sevilla, Spain
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Kristofer Fritz
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Shyam Biswal
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging Intramural Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
28
|
Zhang H, Chen Y, Chen Y, Ji S, Jia P, Xu J, Li Y, Wang T. Pterostilbene attenuates liver injury and oxidative stress in intrauterine growth-retarded weanling piglets. Nutrition 2020; 81:110940. [PMID: 32755743 DOI: 10.1016/j.nut.2020.110940] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/20/2020] [Accepted: 05/27/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES The aim of the present study was to investigate the potential of pterostilbene, a beneficial component primarily found in blueberries, to alleviate the intrauterine growth retardation (IUGR)-induced early liver injury and oxidative stress in a porcine model. METHODS Thirty-six IUGR piglets and an equal number of normal birth weight (NBW) counterparts received a diet with or without pterostilbene (250 mg/kg diet) during the first week post-weaning. Parameters related to the hepatic injury, oxidative stress, and antioxidant defense mechanisms were analyzed. RESULTS Relative to NBW, IUGR induced liver injury, which corresponded to increments in circulating alanine transaminase activity and hepatic apoptotic cell rate, superoxide radical generation, and the accumulation of oxidative damage products (P < 0.05). Administering pterostilbene reduced plasma transaminase activities, decreased hepatocyte apoptosis rate, and prevented the augmented levels of hepatic superoxide anion, 8-hydroxy-2 deoxyguanosine, and 4-hydroxynonenal-modified protein (P < 0.05). In terms of the hepatic antioxidant function, pterostilbene was efficient in improving the superoxide dismutase activity and the metabolic cycle between reduced glutathione and its oxidized form (P < 0.05). The pterostilbene-supplemented diet facilitated the nuclear translocation of nuclear factor erythroid-2-related factor 2 (NRF2) and promoted the expression levels of superoxide dismutase 2 in the liver of IUGR piglets (P < 0.05). CONCLUSION This study indicates that pterostilbene treatment has an auxiliary therapeutic potential to ameliorate early liver injury in IUGR neonates, presumably by stimulating the NRF2 signals and the associated antioxidant function.
Collapse
Affiliation(s)
- Hao Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China; Postdoctoral Research Station of Clinical Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P. R. China; Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai, P. R. China
| | - Yanan Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China
| | - Yueping Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China
| | - Shuli Ji
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China
| | - Peilu Jia
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China
| | - Jianxiong Xu
- Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai, P. R. China; School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Yue Li
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, P. R. China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China.
| |
Collapse
|
29
|
Lipidomic Biomarkers in Polycystic Ovary Syndrome and Endometrial Cancer. Int J Mol Sci 2020; 21:ijms21134753. [PMID: 32635401 PMCID: PMC7370092 DOI: 10.3390/ijms21134753] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Women with polycystic ovary syndrome (PCOS) are more likely to develop endometrial cancer (EC). The molecular mechanisms which increase the risk of EC in PCOS are unclear. Derangements in lipid metabolism are associated with EC, but there have been no studies, investigating if this might increase the risk of EC in PCOS. This was a cross-sectional study of 102 women in three groups of 34 (PCOS, EC and controls) at Nottingham University Hospital, UK. All participants had clinical assessments, followed by obtaining plasma and endometrial tissue samples. Lipidomic analyses were performed using liquid chromatography (LC) coupled with high resolution mass spectrometry (HRMS) and the obtained lipid datasets were screened using standard software and databases. Using multivariate data analysis, there were no common markers found for EC and PCOS. However, on univariate analyses, both PCOS and EC endometrial tissue samples showed a significant decrease in monoacylglycerol 24:0 and capric acid compared to controls. Further studies are required to validate these findings and investigate the potential role of monoacylglycerol 24:0 and capric acid in the link between PCOS with EC.
Collapse
|
30
|
Zhang H, Chen Y, Chen Y, Ji S, Jia P, Li Y, Wang T. Comparison of the protective effects of resveratrol and pterostilbene against intestinal damage and redox imbalance in weanling piglets. J Anim Sci Biotechnol 2020; 11:52. [PMID: 32514341 PMCID: PMC7262760 DOI: 10.1186/s40104-020-00460-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/13/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Evidence indicates that early weaning predisposes piglets to intestinal oxidative stress and increases the risk of intestinal dysfunction; however, there are minimal satisfactory treatment strategies for these conditions. This study investigated the potential of resveratrol and its analog, pterostilbene, as antioxidant protectants for regulating intestinal morphology, barrier function, and redox status among weanling piglets. METHODS A total of 144 piglets were selected at 21 days of age and randomly allocated into one of four treatment groups, each of which included six replicates. Piglets in a sow-reared control group were suckling normally between ages 21 and 28 days, while those in weaned groups were fed a basal diet, supplemented with either 300 mg/kg of resveratrol or with 300 mg/kg of pterostilbene. Parameters associated with intestinal injury and redox status were analyzed at the end of the feeding trial. RESULTS Early weaning disrupted the intestinal function of young piglets, with evidence of increased diamine oxidase activity and D-lactate content in the plasma, shorter villi, an imbalance between cell proliferation and apoptosis, an impaired antioxidant defense system, and severe oxidative damage in the jejunum relative to suckling piglets. Feeding piglets with a resveratrol-supplemented diet partially increased villus height (P = 0.056) and tended to diminish apoptotic cell numbers (P = 0.084) in the jejunum compared with those fed a basal diet. Similarly, these beneficial effects were observed in the pterostilbene-fed piglets. Pterostilbene improved the feed efficiency of weanling piglets between the ages of 21 and 28 days; it also resulted in diminished plasma diamine oxidase activity and D-lactate content relative to untreated weaned piglets (P < 0.05). Notably, pterostilbene restored jejunal antioxidant capacity, an effect that was nearly absent in the resveratrol-fed piglets. Pterostilbene reduced the malondialdehyde and 8-hydroxy-2´-deoxyguanosine contents of jejunal mucosa possibly through its regulatory role in facilitating the nuclear translocation of nuclear factor erythroid-2-related factor 2 and the expression levels of NAD(P)H quinone dehydrogenase 1 and superoxide dismutase 2 (P < 0.05). CONCLUSIONS The results indicate that pterostilbene may be more effective than its parent compound in alleviating early weaning-induced intestinal damage and redox imbalance among young piglets.
Collapse
Affiliation(s)
- Hao Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
- Postdoctoral Research Station of Clinical Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
- Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai, 200240 China
| | - Yanan Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Yueping Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
- Postdoctoral Research Station of Food Science and Engineering, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Shuli Ji
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Peilu Jia
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Yue Li
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014 China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| |
Collapse
|
31
|
Kim S, Hwang JS, Lee DG. Lactoferricin B like peptide triggers mitochondrial disruption‐mediated apoptosis by inhibiting respiration under nitric oxide accumulation in
Candida albicans. IUBMB Life 2020; 72:1515-1527. [DOI: 10.1002/iub.2284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Suhyun Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu South Korea
| | - Jae Sam Hwang
- Department of Agricultural BiologyNational Academy of Agricultural Science, RDA Wanju Republic of Korea
| | - Dong Gun Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu South Korea
| |
Collapse
|
32
|
|
33
|
Mokarizadeh N, Karimi P, Erfani M, Sadigh-Eteghad S, Fathi Maroufi N, Rashtchizadeh N. β-Lapachone attenuates cognitive impairment and neuroinflammation in beta-amyloid induced mouse model of Alzheimer's disease. Int Immunopharmacol 2020; 81:106300. [PMID: 32070922 DOI: 10.1016/j.intimp.2020.106300] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/19/2020] [Accepted: 02/09/2020] [Indexed: 12/11/2022]
Abstract
Oxidative stress and neuroinflammation are critically involved in amyloid beta (Aβ) induced cognitive impairments. β-Lapachone (β-LAP) is a natural activator of NAD(P)H quinone oxidoreductase 1 (NQO1) which has antioxidant and anti-inflammatory properties.This study investigated the effect of β-LAP administration on Aβ-induced memory deficit, oxidative stress, neuroinflammation, and apoptosis cell death in the hippocampus. Forty BALB/c mice were allocated into control, sham, β-LAP (βL), Aβ, and Aβ + βL groups. Intracerebroventricular injection of Aβ1-42 was used to induce Alzheimer's disease (AD) model. Mice in the βL and Aβ + βL groups were treated with β-LAP (10 mg/kg, i.p) for 4 days. Results revealed that β-LAP attenuated memory impairment in the Aβ-received mice, as measured in the novel object recognition (NOR) and Barnes maze tests. Moreover, Aβ resulted in inflammasome activation evident by enhanced caspase-1 immunoreactivity and interleukin-1 beta (IL-1β) protein levels. However, β-LAP could markedly reduce reactive oxygen species (ROS) production and down-regulate mRNA expression of NLRP3 inflammasome and protein levels of cleaved caspase 1 and IL-1β. Additionally, β-LAP-treated mice showed increased SIRT1 levels and NAD+/NADH ratio in the hippocampus. These results were followed by fewer number of TUNEL-positive cell, reduced hippocampal atrophy and neuronal loss in the hippocampal dentate gyrus (DG). These results indicated that the protective effect of β-LAP against AD-associated cognitive deficits is partially through its strong antioxidant and anti-inflammatory actions.
Collapse
Affiliation(s)
- Narmin Mokarizadeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Erfani
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadereh Rashtchizadeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Connective Tissue Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
34
|
Xu F, Wang M, Hu S, Zhou Y, Collyer J, Li K, Xu H, Xiao J. Candidate Regulators of Dyslipidemia in Chromosome 1 Substitution Lines Using Liver Co-Expression Profiling Analysis. Front Genet 2020; 10:1258. [PMID: 31998355 PMCID: PMC6962132 DOI: 10.3389/fgene.2019.01258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/14/2019] [Indexed: 11/13/2022] Open
Abstract
Dyslipidemia is a major risk factor for cardiovascular disease. Although many genetic factors have been unveiled, a large fraction of the phenotypic variance still needs further investigation. Chromosome 1 (Chr 1) harbors multiple gene loci that regulate blood lipid levels, and identifying functional genes in these loci has proved challenging. We constructed a mouse population, Chr 1 substitution lines (C1SLs), where only Chr 1 differs from the recipient strain C57BL/6J (B6), while the remaining chromosomes are unchanged. Therefore, any phenotypic variance between C1SLs and B6 can be attributed to the differences in Chr 1. In this study, we assayed plasma lipid and glucose levels in 13 C1SLs and their recipient strain B6. Through weighted gene co-expression network analysis of liver transcriptome and “guilty-by-association” study, eight associated modules of plasma lipid and glucose were identified. Further joint analysis of human genome wide association studies revealed 48 candidate genes. In addition, 38 genes located on Chr 1 were also uncovered, and 13 of which have been functionally validated in mouse models. These results suggest that C1SLs are ideal mouse models to identify functional genes on Chr 1 associated with complex traits, like dyslipidemia, by using gene co-expression network analysis.
Collapse
Affiliation(s)
- Fuyi Xu
- College of Chemistry, Chemical Engineering, and Biotechnology, Donghua University, Shanghai, China
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Maochun Wang
- College of Chemistry, Chemical Engineering, and Biotechnology, Donghua University, Shanghai, China
| | - Shixian Hu
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Yuxun Zhou
- College of Chemistry, Chemical Engineering, and Biotechnology, Donghua University, Shanghai, China
| | - John Collyer
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kai Li
- College of Chemistry, Chemical Engineering, and Biotechnology, Donghua University, Shanghai, China
| | - Hongyan Xu
- Department of Biostatistics and Epidemiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Junhua Xiao
- College of Chemistry, Chemical Engineering, and Biotechnology, Donghua University, Shanghai, China
- *Correspondence: Junhua Xiao,
| |
Collapse
|
35
|
Katsyuba E, Romani M, Hofer D, Auwerx J. NAD + homeostasis in health and disease. Nat Metab 2020; 2:9-31. [PMID: 32694684 DOI: 10.1038/s42255-019-0161-5] [Citation(s) in RCA: 369] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022]
Abstract
The conceptual evolution of nicotinamide adenine dinucleotide (NAD+) from being seen as a simple metabolic cofactor to a pivotal cosubstrate for proteins regulating metabolism and longevity, including the sirtuin family of protein deacylases, has led to a new wave of scientific interest in NAD+. NAD+ levels decline during ageing, and alterations in NAD+ homeostasis can be found in virtually all age-related diseases, including neurodegeneration, diabetes and cancer. In preclinical settings, various strategies to increase NAD+ levels have shown beneficial effects, thus starting a competitive race to discover marketable NAD+ boosters to improve healthspan and lifespan. Here, we review the basics of NAD+ biochemistry and metabolism, and its roles in health and disease, and we discuss current challenges and the future translational potential of NAD+ research.
Collapse
Affiliation(s)
- Elena Katsyuba
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Nagi Bioscience, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mario Romani
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Dina Hofer
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Thermo Fisher Scientific, Zug, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
36
|
Diaz-Ruiz A, Di Francesco A, Carboneau BA, Levan SR, Pearson KJ, Price NL, Ward TM, Bernier M, de Cabo R, Mercken EM. Benefits of Caloric Restriction in Longevity and Chemical-Induced Tumorigenesis Are Transmitted Independent of NQO1. J Gerontol A Biol Sci Med Sci 2019; 74:155-162. [PMID: 29733330 DOI: 10.1093/gerona/gly112] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/02/2018] [Indexed: 12/22/2022] Open
Abstract
Caloric restriction (CR) is the most potent nonpharmacological intervention known to both protect against carcinogenesis and delay aging in laboratory animals. There is a growing number of anticarcinogens and CR mimetics that activate NAD(P)H:quinone oxidoreductase 1 (NQO1). We have previously shown that NQO1, an antioxidant enzyme that acts as an energy sensor through modulation of intracellular redox and metabolic state, is upregulated by CR. Here, we used NQO1-knockout (KO) mice to investigate the role of NQO1 in both the aging process and tumor susceptibility, specifically in the context of CR. We found that NQO1 is not essential for the beneficial effects of CR on glucose homeostasis, physical performance, metabolic flexibility, life-span extension, and (unlike our previously observation with Nrf2) chemical-induced tumorigenesis.
Collapse
Affiliation(s)
- Alberto Diaz-Ruiz
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland.,Nutritional Interventions Group, Precision Nutrition and Aging, Institute IMDEA Food, Madrid, Spain
| | - Andrea Di Francesco
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Bethany A Carboneau
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Sophia R Levan
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Kevin J Pearson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington
| | - Nathan L Price
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT
| | - Theresa M Ward
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland.,Nutritional Interventions Group, Precision Nutrition and Aging, Institute IMDEA Food, Madrid, Spain
| | - Evi M Mercken
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
37
|
Zhu FC, Jiang DM, Zhang MH, Zhao B, He C, Yang J. Adenovirus vector‑mediated in vivo gene transfer of nuclear factor erythroid‑2p45‑related factor 2 promotes functional recovery following spinal cord contusion. Mol Med Rep 2019; 20:4285-4292. [PMID: 31545436 DOI: 10.3892/mmr.2019.10687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 07/24/2019] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate whether nuclear factor erythroid 2p45‑related factor 2 (Nrf2) overexpression by gene transfer may protect neurons/glial cells and the association between neurons/glial cells and axons in spinal cord injury (SCI). In the present study, Nrf2 recombinant adenovirus (Ad) vectors were constructed. The protein levels of Nrf2 in the nucleus and of the Nrf2‑regulated gene products heme oxygenase‑1 (HO‑1) and NAD (P)H‑quinone oxidoreductase‑1 (NQO1), were detected using western blot analysis in PC12 cells following 48 h of transfection. Furthermore, the expression of Nrf2 was localized using an immunofluorescence experiment, and the expression of Nrf2, HO‑1 and NQO1 were detected using an immunohistochemical experiment in the grey matter of spinal cord in rats. Post‑injury motor behavior was assessed via the Basso, Beattie and Bresnahan (BBB) locomotor scale method. In PC12 cells, subsequent to Ad‑Nrf2 transfection, nuclear Nrf2, HO‑1 and NQO1 levels were significantly increased compared with the control (P<0.01). There was statistically significant changes in the PC12‑Ad‑Nrf2 group [Nrf2 (1.146±0.095), HO‑1 (1.816±0.095) and NQO1 (1.421±0.138)] compared with the PC12‑control group [Nrf2 (0.717±0.055), HO‑1 (1.264±0.081) and NQO1 (0.921±0.088)] and PC12‑Ad‑green fluorescent protein group [Nrf2 (0.714±0.111), HO‑1 (1.238±0.053) and NQO1 (0.987±0.045); P<0.01]. The BBB scores of the rats indicated that they had improved functional recovery following the local injection of Ad‑Nrf2. On the third day following the operation, BBB scores in the adenovirus groups (0.167±0.408) were significantly decreased compared with the SCI group (1±0.894; P<0.05). In the injured section of the spinal cord in the rats, the number of positive cells expressing Nrf2, HO‑1 and NQO1 were raised compared with the control and SCI groups, indicating that the adenovirus vector‑mediated gene transfer of Nrf2 promotes functional recovery following spinal cord contusion in rats.
Collapse
Affiliation(s)
- Feng-Chen Zhu
- Department of Orthopaedics, Yongchuan Affiliated Hospital, Chongqing Medical University, Chongqing 402160, P.R. China
| | - Dian-Ming Jiang
- Department of Orthopaedics, The Third Affiliated Hospital, Chongqing Medical University, Chongqing 401120, P.R. China
| | - Ming-Hua Zhang
- Department of Orthopaedics, Yongchuan Affiliated Hospital, Chongqing Medical University, Chongqing 402160, P.R. China
| | - Bo Zhao
- Department of Orthopaedics, Yongchuan Affiliated Hospital, Chongqing Medical University, Chongqing 402160, P.R. China
| | - Chao He
- Department of Orthopaedics, Yongchuan Affiliated Hospital, Chongqing Medical University, Chongqing 402160, P.R. China
| | - Jian Yang
- Department of Orthopaedics, Yongchuan Affiliated Hospital, Chongqing Medical University, Chongqing 402160, P.R. China
| |
Collapse
|
38
|
Kim CY, Kang B, Hong J, Choi HS. Parthenolide inhibits lipid accumulation via activation of Nrf2/Keap1 signaling during adipocyte differentiation. Food Sci Biotechnol 2019; 29:431-440. [PMID: 32257527 DOI: 10.1007/s10068-019-00672-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/04/2019] [Accepted: 08/13/2019] [Indexed: 12/22/2022] Open
Abstract
The effects of parthenolide (PL), a sesquiterpene lactone obtained from feverfew plant, on lipid accumulation and signaling pathway in adipocytes were investigated. PL significantly inhibited lipid accumulation and adipogenic factors during adipogenesis. In particular, PL exerted its inhibitory effects in early adipogenic stage by regulating the early adipogenic factors. In addition, PL regulated the expression of adipokines; leptin, retinol binding protein, and resistin mRNAs were downregulated, whereas adiponectin gene expression was increased. Furthermore, PL significantly reduced intracellular reactive oxygen species (ROS) production during adipogenesis. This PL-mediated regulation of ROS production was associated with the regulation of nuclear factor erythroid 2-related factor (Nrf2)-kelch-like ECH-associated protein 1 (Keap1) pathway. PL effectively increased the abundance of Nrf2 and its target proteins, heme oxygenase-1 (HO-1) and NADPH dehydrogenase 1 (NQO1), by promoting the nuclear translocation of Nrf2, indicating that PL-mediated anti-adipogenic effects are associated with the Nrf2/Keap1 pathway.
Collapse
Affiliation(s)
- Chae Young Kim
- 1Department of Public Health Sciences, Korea University, Seoul, 07249 Republic of Korea
| | - Bobin Kang
- 1Department of Public Health Sciences, Korea University, Seoul, 07249 Republic of Korea
| | - Jungil Hong
- 2Department of Food Science and Technology, College of Natural Science, Seoul Women's University, Seoul, 139-774 Republic of Korea
| | - Hyeon-Son Choi
- 2Department of Food Science and Technology, College of Natural Science, Seoul Women's University, Seoul, 139-774 Republic of Korea
| |
Collapse
|
39
|
Yin and Yang: Why did evolution implement and preserve the circadian rhythmicity? Med Hypotheses 2019; 131:109306. [PMID: 31443763 DOI: 10.1016/j.mehy.2019.109306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/15/2019] [Accepted: 07/06/2019] [Indexed: 02/07/2023]
Abstract
Yin and Yang concept emphasizes the reciprocal and interrelated nature; neither is sufficient, both are needed to sustain the overall balance of the living system. Changing the balance, by implementing deficiency or excess of one of them, upsets the equilibrium (homeostasis) of the whole system. PURPOSE In this opinion article intermittent exposure is presented as the stimulus for development and evolutionary conservation of circadian rhythm, an endogenous, entrainable oscillation of approximately 24 h, to counteract/balance the cells' natural tendency to attenuate their response during long-term exposure to different endogenous substances. RESULTS The concept of Yin and Yang duality is an allegory on which the avoidance of attenuation of the cells' responses hypothesis is presented as an explanation for the circadian rhythmicity, which is integrated in all human cells, with the exception of stem and cancer cells. CONCLUSIONS We hypothesize, that circadian rhythmicity has evolved, during evolution, into a mechanism that prevents disruption of the organism's negative-feedback-loop homeostasis.
Collapse
|
40
|
Punganuru SR, Madala HR, Arutla V, Zhang R, Srivenugopal KS. Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging. Sci Rep 2019; 9:8577. [PMID: 31189950 PMCID: PMC6562040 DOI: 10.1038/s41598-019-44111-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/09/2019] [Indexed: 12/30/2022] Open
Abstract
The Near-infrared Fluorescence (NIRF) molecular imaging of cancer is known to be superior in sensitivity, deeper penetration, and low phototoxicity compared to other imaging modalities. In view of an increased need for efficient and targeted imaging agents, we synthesized a NAD(P)H quinone oxidoreductase 1 (NQO1)-activatable NIR fluorescent probe (NIR-ASM) by conjugating dicyanoisophorone (ASM) fluorophore with the NQO1 substrate quinone propionic acid (QPA). The probe remained non-fluorescent until activation by NQO1, whose expression is largely limited to malignant tissues. With a large Stokes shift (186 nm) and a prominent near-infrared emission (646 nm) in response to NQO1, NIR-ASM was capable of monitoring NQO1 activity in vitro and in vivo with high specificity and selectivity. We successfully employed the NIR-ASM to differentiate cancer cells from normal cells based on NQO1 activity using fluorescence microscopy and flow cytometry. Chemical and genetic approaches involving the use of ES936, a specific inhibitor of NQO1 and siRNA and gene transfection procedures unambiguously demonstrated NQO1 to be the sole target activating the NIR-ASM in cell cultures. NIR-ASM was successfully used to detect and image the endogenous NQO1 in three live tumor-bearing mouse models (A549 lung cancer, Lewis lung carcinoma, and MDMAMB 231 xenografts) with a high signal-to-low noise ratiometric NIR fluorescence response. When the NQO1-proficient A549 tumors and NQO1-deficient MDA-MB-231 tumors were developed in the same animal, only the A549 malignancies activated the NIR-ASM probe with a strong signal. Because of its high sensitivity, rapid activation, tumor selectivity, and nontoxic properties, the NIR-ASM appears to be a promising agent with clinical applications.
Collapse
Affiliation(s)
- Surendra Reddy Punganuru
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| | - Hanumantha Rao Madala
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Viswanath Arutla
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA
| | - Kalkunte S Srivenugopal
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| |
Collapse
|
41
|
Calvo-Garrido J, Maffezzini C, Schober FA, Clemente P, Uhlin E, Kele M, Stranneheim H, Lesko N, Bruhn H, Svenningsson P, Falk A, Wedell A, Freyer C, Wredenberg A. SQSTM1/p62-Directed Metabolic Reprogramming Is Essential for Normal Neurodifferentiation. Stem Cell Reports 2019; 12:696-711. [PMID: 30827875 PMCID: PMC6449840 DOI: 10.1016/j.stemcr.2019.01.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 02/02/2023] Open
Abstract
Neurodegenerative disorders are an increasingly common and irreversible burden on society, often affecting the aging population, but their etiology and disease mechanisms are poorly understood. Studying monogenic neurodegenerative diseases with known genetic cause provides an opportunity to understand cellular mechanisms also affected in more complex disorders. We recently reported that loss-of-function mutations in the autophagy adaptor protein SQSTM1/p62 lead to a slowly progressive neurodegenerative disease presenting in childhood. To further elucidate the neuronal involvement, we studied the cellular consequences of loss of p62 in a neuroepithelial stem cell (NESC) model and differentiated neurons derived from reprogrammed p62 patient cells or by CRISPR/Cas9-directed gene editing in NESCs. Transcriptomic and proteomic analyses suggest that p62 is essential for neuronal differentiation by controlling the metabolic shift from aerobic glycolysis to oxidative phosphorylation required for neuronal maturation. This shift is blocked by the failure to sufficiently downregulate lactate dehydrogenase expression due to the loss of p62, possibly through impaired Hif-1α downregulation and increased sensitivity to oxidative stress. The findings imply an important role for p62 in neuronal energy metabolism and particularly in the regulation of the shift between glycolysis and oxidative phosphorylation required for normal neurodifferentiation.
Collapse
Affiliation(s)
- Javier Calvo-Garrido
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Camilla Maffezzini
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Florian A Schober
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Paula Clemente
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Elias Uhlin
- Department of Neuroscience, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Malin Kele
- Department of Neuroscience, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Henrik Stranneheim
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Nicole Lesko
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Helene Bruhn
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Anna Wedell
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden; Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Christoph Freyer
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| | - Anna Wredenberg
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| |
Collapse
|
42
|
Ansari MN, Aloliet RI, Ganaie MA, Khan TH, Najeeb-ur-Rehman, Imam F, Hamad AM. Roflumilast, a phosphodiesterase 4 inhibitor, attenuates cadmium-induced renal toxicity via modulation of NF-κB activation and induction of NQO1 in rats. Hum Exp Toxicol 2019; 38:588-597. [DOI: 10.1177/0960327119829521] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: In the present study, the protective effect of Roflumilast (ROF, a selective phosphodiesterase (PDE-4) inhibitor) was investigated against cadmium (Cd)-induced nephrotoxicity in rats. Methods: A total of 24 rats were selected and randomly divided into four groups ( n = 6). Group 1 served as the control; groups 2–4 administered with CdCl2 (3 mg/kg, i.p.) for 7 days; groups 3 and 4 were co-administered with ROF in doses of 0.5 and 1.5 mg/kg, orally for 7 consecutive days. Nephrotoxicity was evaluated by measuring urine volume, urea and creatinine levels in urine and serum. Oxidative stress was confirmed by measuring malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT) levels in kidney tissue followed by histopathological studies. Results: CdCl2 administration results in a significant ( p < 0.01) decrease in urine volume, urea, and creatinine levels in urine, as well as GSH, SOD, and CAT levels in renal tissue. In addition, Cd also produced significantly increased ( p < 0.01) urea and creatinine levels in serum and TBARS levels in renal tissues. Rats treated with ROF significantly ( p < 0.01) restore the altered levels of kidney injury markers, nonenzymatic antioxidant, as well as depleted enzymes in dose-dependent manner. An increased expression of NF-κB p65 and decreased expression of GST and NQO1 in the Cd only treated group were significantly reversed by high dose of ROF (1.5 mg/kg). Histopathological changes were also ameliorated by ROF administration in Cd-treated groups. Conclusion: In conclusion, ROF treatment showed protective effect against renal damage and increased oxidative stress induced by Cd administration.
Collapse
Affiliation(s)
- MN Ansari
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - RI Aloliet
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - MA Ganaie
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - TH Khan
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - Najeeb-ur-Rehman
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - F Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - AM Hamad
- Department of Basic Sciences, Preparatory year Deanship, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
- Department of Histopathology and Cytopathology, Faculty of Medical Laboratory Sciences, University of Gezira, Wad Madani, Sudan
| |
Collapse
|
43
|
Hananya N, Reid JP, Green O, Sigman MS, Shabat D. Rapid chemiexcitation of phenoxy-dioxetane luminophores yields ultrasensitive chemiluminescence assays. Chem Sci 2019; 10:1380-1385. [PMID: 30809354 PMCID: PMC6354826 DOI: 10.1039/c8sc04280b] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/12/2018] [Indexed: 11/21/2022] Open
Abstract
The utility of dioxetane-based chemiluminescent probes in biosensing and bioimaging is being increasingly recognized. While phenoxy-dioxetane luminophores with fast chemiexcitation kinetics are highly desired, current luminophores suffer from slow chemiexcitation. Herein we describe a rational, computationally-supported design of phenoxy-dioxetanes with fast chemiexcitation kinetics. These new luminophores were designed to contain a substituent that promotes rapid chemiexcitation, emitting light up to 100-fold faster than currently known dioxetanes. We demonstrate the superiority of the new phenoxy-dioxetanes by preparing three chemiluminescent probes for NAD(P)H, which differ from each other in the rate of the chemiexcitation. Comparison of these probes reveals a correlation between the chemiexcitation rate and the probe sensitivity. We anticipate that these new phenoxy-dioxetanes could serve as an ideal platform for designing chemiluminescence probes with enhanced sensitivity for numerous bioassays.
Collapse
Affiliation(s)
- Nir Hananya
- School of Chemistry , Faculty of Exact Sciences , Tel Aviv University , Tel Aviv 69978 , Israel .
| | - Jolene P Reid
- Department of Chemistry , University of Utah , 315 South 1400 East , Salt Lake City , Utah 84112 , USA .
| | - Ori Green
- School of Chemistry , Faculty of Exact Sciences , Tel Aviv University , Tel Aviv 69978 , Israel .
| | - Matthew S Sigman
- Department of Chemistry , University of Utah , 315 South 1400 East , Salt Lake City , Utah 84112 , USA .
| | - Doron Shabat
- School of Chemistry , Faculty of Exact Sciences , Tel Aviv University , Tel Aviv 69978 , Israel .
| |
Collapse
|
44
|
Mattioli R, Francioso A, d'Erme M, Trovato M, Mancini P, Piacentini L, Casale AM, Wessjohann L, Gazzino R, Costantino P, Mosca L. Anti-Inflammatory Activity of A Polyphenolic Extract from Arabidopsis thaliana in In Vitro and In Vivo Models of Alzheimer's Disease. Int J Mol Sci 2019; 20:ijms20030708. [PMID: 30736391 PMCID: PMC6387160 DOI: 10.3390/ijms20030708] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/01/2019] [Accepted: 02/03/2019] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder and the primary form of dementia in the elderly. One of the main features of AD is the increase in amyloid-beta (Aβ) peptide production and aggregation, leading to oxidative stress, neuroinflammation and neurodegeneration. Polyphenols are well known for their antioxidant, anti-inflammatory and neuroprotective effects and have been proposed as possible therapeutic agents against AD. Here, we investigated the effects of a polyphenolic extract of Arabidopsis thaliana (a plant belonging to the Brassicaceae family) on inflammatory response induced by Aβ. BV2 murine microglia cells treated with both Aβ25–35 peptide and extract showed a lower pro-inflammatory (IL-6, IL-1β, TNF-α) and a higher anti-inflammatory (IL-4, IL-10, IL-13) cytokine production compared to cells treated with Aβ only. The activation of the Nrf2-antioxidant response element signaling pathway in treated cells resulted in the upregulation of heme oxygenase-1 mRNA and in an increase of NAD(P)H:quinone oxidoreductase 1 activity. To establish whether the extract is also effective against Aβ-induced neurotoxicity in vivo, we evaluated its effect on the impaired climbing ability of AD Drosophila flies expressing human Aβ1–42. Arabidopsis extract significantly restored the locomotor activity of these flies, thus confirming its neuroprotective effects also in vivo. These results point to a protective effect of the Arabidopsis extract in AD, and prompt its use as a model in studying the impact of complex mixtures derived from plant-based food on neurodegenerative diseases.
Collapse
Affiliation(s)
- Roberto Mattioli
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Roma, Italy.
| | - Antonio Francioso
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Roma, Italy.
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06120 Halle (Saale), Germany.
| | - Maria d'Erme
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Roma, Italy.
| | - Maurizio Trovato
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Roma, Italy.
| | - Patrizia Mancini
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Roma, Italy.
| | - Lucia Piacentini
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Roma, Italy.
| | - Assunta Maria Casale
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Roma, Italy.
| | - Ludger Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06120 Halle (Saale), Germany.
| | - Roberta Gazzino
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Roma, Italy.
| | - Paolo Costantino
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Roma, Italy.
| | - Luciana Mosca
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Roma, Italy.
| |
Collapse
|
45
|
New drugs for pharmacological extension of replicative life span in normal and progeroid cells. NPJ Aging Mech Dis 2019; 5:2. [PMID: 30675378 PMCID: PMC6335401 DOI: 10.1038/s41514-018-0032-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
A high-throughput anti-aging drug screen was developed that simultaneously measures senescence-associated β-galactosidase activity and proliferation. Applied to replicatively pre-aged fibroblasts, this screen yielded violuric acid (VA) and 1-naphthoquinone-2-monoxime (N2N1) as its top two hits. These lead compounds extended the replicative life spans of normal and progeroid human cells in a dose-dependent manner and also extended the chronological life spans of mice and C. elegans. They are further shown here to function as redox catalysts in oxidations of NAD(P)H. They thus slow age-related declines in NAD(P)+/NAD(P)H ratios. VA participates in non-enzymatic electron transfers from NAD(P)H to oxidized glutathione or peroxides. N2N1 transfers electrons from NAD(P)H to cytochrome c or CoQ10 via NAD(P)H dehydrogenase (quinone) 1 (NQO1). Our results indicate that pharmacologic manipulation of NQO1 activity via redox catalysts may reveal mechanisms of senescence and aging. Two drugs were discovered that can extend the life spans of normally aged human cells and thus potentially slow human aging. The anti-aging drugs were identified using a novel method that screens drugs across a two-dimensional endpoint space of senescence-associated galactosidase activity as a general axis of aging and ATP as an axis representing proliferation. The two most potent substances were, likely more than coincidentally, electrons carriers that transfer electrons from NAD(P)H to molecules and cellular structures that demand reducing power to repair oxidative damage that accumulates with aging. Treatment of single cells and whole organisms with these new anti-aging drugs increased their lifespans. The mechanism of the drug action may advance our understanding of the complex, yet resolvable, biological process of aging.
Collapse
|
46
|
Kitakaze T, Makiyama A, Samukawa Y, Jiang S, Yamashita Y, Ashida H. A physiological concentration of luteolin induces phase II drug-metabolizing enzymes through the ERK1/2 signaling pathway in HepG2 cells. Arch Biochem Biophys 2019; 663:151-159. [PMID: 30641047 DOI: 10.1016/j.abb.2019.01.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/06/2019] [Accepted: 01/10/2019] [Indexed: 12/20/2022]
Abstract
The flavon luteolin has various health-promoting activities including cardiovascular protection, anti-inflammatory activity and anticancer activity. A serum concentration of about 100 nM luteolin is reached by dietary habit. However, little is known about the function of luteolin over its physiological concentration range. In this study, we investigated whether a physiological concentration of luteolin could activate nuclear factor-erythroid-2-related factor 2 (Nrf2)-mediated expression of phase II drug-metabolizing enzymes in human hepatoma HepG2 cells. Interestingly, less than 1 nM of luteolin could induce phase II drug-metabolizing enzymes, such as GSTs, HO-1, and NQO1. Both 1 and 100 nM luteolin increased expression and activity of ALDH2, which metabolized toxic acetaldehyde into nontoxic acetic acid. Luteolin increased nuclear accumulation of Nrf2 and enhanced the ARE-binding complex through increasing the stability of the Nrf2 protein. Luteolin increased phosphorylation of Nrf2 at Ser40, and MEK inhibitors (U0126 and PD98059) canceled luteolin-induced phosphorylation of Nrf2. Furthermore, luteolin increased modified Keap1. In conclusion, a physiological concentration of luteolin induces the expression of phase II drug-metabolizing enzymes by enhancement of Nrf2 nuclear accumulation through MEK1/2-ERK1/2-mediated phosphorylation of Nrf2, increasing Nrf2 stability and inducing a conformational change of Keap1.
Collapse
Affiliation(s)
- Tomoya Kitakaze
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Hyogo, Japan
| | - Atsushi Makiyama
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | - Yumi Samukawa
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | - Songyan Jiang
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | - Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan.
| |
Collapse
|
47
|
Molano-Arevalo JC, Gonzalez W, Jeanne Dit Fouque K, Miksovska J, Maitre P, Fernandez-Lima F. Insights from ion mobility-mass spectrometry, infrared spectroscopy, and molecular dynamics simulations on nicotinamide adenine dinucleotide structural dynamics: NAD +vs. NADH. Phys Chem Chem Phys 2018; 20:7043-7052. [PMID: 29473073 DOI: 10.1039/c7cp05602h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD) is found in all living cells where the oxidized (NAD+) and reduced (NADH) forms play important roles in many enzymatic reactions. However, little is known about NAD+ and NADH conformational changes and kinetics as a function of the cell environment. In the present work, an analytical workflow is utilized to study NAD+ and NADH dynamics as a function of the organic content in solution using fluorescence lifetime spectroscopy and in the gas-phase using trapped ion mobility spectrometry coupled to mass spectrometry (TIMS-MS) and infrared multiple photon dissociation (IRMPD) spectroscopy. NAD solution time decay studies showed a two-component distribution, assigned to changes from a "close" to "open" conformation with the increase of the organic content. NAD gas-phase studies using nESI-TIMS-MS displayed two ion mobility bands for NAD+ protonated and sodiated species, while four and two ion mobility bands were observed for NADH protonated and sodiated species, respectively. Changes in the mobility profiles were observed for NADH as a function of the starting solution conditions and the time after desolvation, while NAD+ profiles showed no dependence. IRMPD spectroscopy of NAD+ and NADH protonated species in the 800-1800 and 3200-3700 cm-1 spectral regions showed common and signature bands between the NAD forms. Candidate structures were proposed for NAD+ and NADH kinetically trapped intermediates of the protonated and sodiated species, based on their collision cross sections and IR profiles. Results showed that NAD+ and NADH species exist in open, stack, and closed conformations and that the driving force for conformational dynamics is hydrogen bonding of the N-H-O and O-H-O forms with ribose rings.
Collapse
|
48
|
Khadka D, Kim HJ, Oh GS, Shen A, Lee S, Lee SB, Sharma S, Kim SY, Pandit A, Choe SK, Kwak TH, Yang SH, Sim H, Eom GH, Park R, So HS. Augmentation of NAD + levels by enzymatic action of NAD(P)H quinone oxidoreductase 1 attenuates adriamycin-induced cardiac dysfunction in mice. J Mol Cell Cardiol 2018; 124:45-57. [PMID: 30291911 DOI: 10.1016/j.yjmcc.2018.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/18/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Adriamycin (ADR) is a powerful chemotherapeutic agent extensively used to treat various human neoplasms. However, its clinical utility is hampered due to severe adverse side effects i.e. cardiotoxicity and heart failure. ADR-induced cardiomyopathy (AIC) has been reported to be caused by myocardial damage and dysfunction through oxidative stress, DNA damage, and inflammatory responses. Nonetheless, the remedies for AIC are even not established. Therefore, we illustrate the role of NAD+/NADH modulation by NAD(P)H quinone oxidoreductase 1 (NQO1) enzymatic action on AIC. METHODS AND RESULTS AIC was established by intraperitoneal injection of ADR in C57BL/6 wild-type (WT) and NQO1 knockout (NQO1-/-) mice. All Mice were orally administered dunnione (named NQO1 substrate) before and after exposure to ADR. Cardiac biomarker levels in the plasma, cardiac dysfunction, oxidative biomarkers, and mRNA and protein levels of pro-inflammatory mediators were determined compared the cardiac toxicity of each experimental group. All biomarkers of Cardiac damage and oxidative stress, and mRNA levels of pro-inflammatory cytokines including cardiac dysfunction were increased in ADR-treated both WT and NQO1-/- mice. However, this increase was significantly reduced by dunnione in WT, but not in NQO1-/- mice. In addition, a decrease in SIRT1 activity due to a reduction in the NAD+/NADH ratio by PARP-1 hyperactivation was associated with AIC through increased nuclear factor (NF)-κB p65 and p53 acetylation in both WT and NQO1-/- mice. While an elevation in NAD+/NADH ratio via NQO1 enzymatic action using dunnione recovered SIRT1 activity and subsequently deacetylated NF-κB p65 and p53, however not in NQO1-/- mice, thereby attenuating AIC. CONCLUSION Thus, modulation of NAD+/NADH by NQO1 may be a novel therapeutic approach to prevent chemotherapy-associated heart failure, including AIC.
Collapse
Affiliation(s)
- Dipendra Khadka
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Hyung-Jin Kim
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Gi-Su Oh
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - AiHua Shen
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - SeungHoon Lee
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Su-Bin Lee
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Subham Sharma
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Seon Young Kim
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Arpana Pandit
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Seong-Kyu Choe
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Tae Hwan Kwak
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Sei-Hoon Yang
- Internal Medicine, School of Medicine Wonkwang, University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Hyuk Sim
- Internal Medicine, School of Medicine Wonkwang, University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Gwang Hyeon Eom
- Department of Pharmacology, Medical Research Center for Gene Regulation Chonnam, National University Medical School, Hwasungun Jeollanam-do 58128, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science & Engineering, Institute of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea..
| |
Collapse
|
49
|
Kang HJ, Song HY, Ahmed MA, Guo Y, Zhang M, Chen C, Cristofanilli M, Horiuchi D, Vassilopoulos A. NQO1 regulates mitotic progression and response to mitotic stress through modulating SIRT2 activity. Free Radic Biol Med 2018; 126:358-371. [PMID: 30114477 PMCID: PMC6170003 DOI: 10.1016/j.freeradbiomed.2018.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 12/30/2022]
Abstract
Previous studies have shown that SIRT2 plays a role in mitosis through deacetylating specific downstream targets. However, the upstream regulation of SIRT2 activity has been relatively unexplored. In this study, we provide evidence that NAD(P)H:quinone oxidoreductase 1 (NQO1) interacts with and activates SIRT2 in an NAD-dependent manner. Strong protein-protein interaction and co-localization of the two proteins during mitosis is required to maintain an active NQO1-SIRT2 axis which is critical for successful completion of mitosis. This is evident by the observed delay in mitotic exit in cells upon NQO1 inhibition. Mechanistically, this phenotype can be explained by the decrease in APC/C complex activity resulting from decreased SIRT2 deacetylation activity. Furthermore, we show that this newly established role of NQO1 has an impact on how cancer cells may respond to mitotic stress. In this regard, both pharmacologic and genetic NQO1 inhibition increases sensitivity to anti-mitotic drugs functioning as microtubule poisons by inducing mitotic arrest and allowing cells to accumulate cell death signals. Therefore, the significant prognostic value of NQO1 in predicting outcome of cancer patients might be explained in part due to the functional contribution of NQO1-SIRT2 axis to mitotic stress. Altogether, this novel mechanism of action further supports the pleiotropic biological effects exerted by NQO1 in addition to its antioxidant function and it might provide the basis for expanding the therapeutic potential of NQO1 inhibition towards increasing sensitivity to standard treatments.
Collapse
Affiliation(s)
- Hong-Jun Kang
- Laboratory for Molecular Cancer Biology, Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ha Yong Song
- Laboratory for Molecular Cancer Biology, Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Mohamed A Ahmed
- Laboratory for Molecular Cancer Biology, Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Radiation Biology Department, National Center for Radiation Research and Technology, Cairo, Egypt
| | - Yang Guo
- Laboratory for Molecular Cancer Biology, Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mingming Zhang
- Laboratory for Molecular Cancer Biology, Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chuyu Chen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Massimo Cristofanilli
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Medicine-Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dai Horiuchi
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Athanassios Vassilopoulos
- Laboratory for Molecular Cancer Biology, Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
50
|
Micheli L, Mattoli L, Maidecchi A, Pacini A, Ghelardini C, Di Cesare Mannelli L. Effect of Vitis vinifera hydroalcoholic extract against oxaliplatin neurotoxicity: in vitro and in vivo evidence. Sci Rep 2018; 8:14364. [PMID: 30254294 PMCID: PMC6156221 DOI: 10.1038/s41598-018-32691-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023] Open
Abstract
Oxaliplatin treatment is associated with the development of a dose-limiting painful neuropathy impairing patient's quality of life. Since oxidative unbalance is a relevant mechanism of oxaliplatin neurotoxicity, we assessed the potential antioxidant properties of Vitis vinifera extract in reducing oxaliplatin-induced neuropathy as a valuable therapeutic opportunity. A hydroalcoholic extract of Vitis vinifera red leaf was characterized and tested in primary rat astrocyte cells treated with oxaliplatin (100 μM). Oxaliplatin lethality in the human adenocarcinoma cell line HT-29 was evaluated in the absence and presence of the extract. In vivo, pain hypersensitivity was measured in a rat model of neuropathy induced by oxaliplatin and ex vivo molecular targets of redox balance were studied. Vitis vinifera extract (50 μg mL-1, 4 h incubation) significantly reduced the oxaliplatin-dependent superoxide anion increase and lipid peroxidation in rat astrocytes but did not interfere with the mortality elicited by oxaliplatin in HT-29 cancer cells. In oxaliplatin-treated rats, a repeated daily administration of the Vitis vinifera extract (300 mg kg-1, p.o.) significantly prevented mechanical and thermal hypersensitivity to noxious and non noxious stimuli. mRNA and protein levels of Nrf2 were normalized in spinal cord and DRGs. Moreover, in the spinal cord, the extract significantly decreased the activation of astrocytes. Vitis vinifera reduced oxidative damages and relieved pain without influencing oxaliplatin anti-cancer activity.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Viale Gaetano Pieraccini 6, 50139, Italy
| | - Luisa Mattoli
- Aboca S.p.A. Società Agricola, Località Aboca, Sansepolcro, Arezzo, 52100, Italy
| | - Anna Maidecchi
- Aboca S.p.A. Società Agricola, Località Aboca, Sansepolcro, Arezzo, 52100, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Florence, Largo Brambilla 1, 50134, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Viale Gaetano Pieraccini 6, 50139, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Viale Gaetano Pieraccini 6, 50139, Italy.
| |
Collapse
|