1
|
Ni L, Jiang C, Guo Q, Chi H, Fan C, Shi J, Lin N, Liu Z, Chen S. Metabolomics analysis of physicochemical properties associated with freshness degradation in frozen Antarctic krill (Euphausia superba). Food Res Int 2024; 183:114190. [PMID: 38760127 DOI: 10.1016/j.foodres.2024.114190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 05/19/2024]
Abstract
This study aimed to determine the effect of different frozen temperatures during storage on the quality of Antarctic krill (Euphausia superba) and assess the change at the metabolite level via a combination of physicochemical property analysis, liquid chromatography-tandem mass spectrometry (LC-MS) based non-targeted metabolomics profiling. Regarding samples stored at -20 °C, the expressions of 7055 metabolites were elevated, while 2313 were downregulated. Lipids and lipid molecules had the highest proportion of differential metabolites. A total of 432 discriminatory metabolites with Kyoto Encyclopedia of Genes and Genomes (KEGG) IDs was obtained. We also observed that the concentrations of differential bitter free amino acids (FAAs) and oxidation products of arachidonic and linoleic acid increased. Moreover, as the storage temperature increased, the freshness, umami, and sweetness components were considerably reduced. Furthermore, results indicated that the color, pH and water-holding capacity (WHC) were potential indicators of quality deterioration, while inosinic acid was a probable biomarker for umami degradation of frozen Antarctic krill. In conclusion, this study demonstrates that storage at lower temperatures can be beneficial for maintaining the freshness of Antarctic krill from macro and micro perspectives.
Collapse
Affiliation(s)
- Ling Ni
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Chaojun Jiang
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Quanyou Guo
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Hai Chi
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Chengqi Fan
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Jiangao Shi
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Na Lin
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Zhidong Liu
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China.
| | - Shengjun Chen
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, National Research and Development Center for Aquatic Product Processing, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China.
| |
Collapse
|
2
|
Wu HHL, McDonnell T, Chinnadurai R. Physiological Associations between Vitamin B Deficiency and Diabetic Kidney Disease. Biomedicines 2023; 11:biomedicines11041153. [PMID: 37189771 DOI: 10.3390/biomedicines11041153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
The number of people living with chronic kidney disease (CKD) is growing as our global population continues to expand. With aging, diabetes, and cardiovascular disease being major harbingers of kidney disease, the number of people diagnosed with diabetic kidney disease (DKD) has grown concurrently. Poor clinical outcomes in DKD could be influenced by an array of factors-inadequate glycemic control, obesity, metabolic acidosis, anemia, cellular senescence, infection and inflammation, cognitive impairment, reduced physical exercise threshold, and, importantly, malnutrition contributing to protein-energy wasting, sarcopenia, and frailty. Amongst the various causes of malnutrition in DKD, the metabolic mechanisms of vitamin B (B1 (Thiamine), B2 (Riboflavin), B3 (Niacin/Nicotinamide), B5 (Pantothenic Acid), B6 (Pyridoxine), B8 (Biotin), B9 (Folate), and B12 (Cobalamin)) deficiency and its clinical impact has garnered greater scientific interest over the past decade. There remains extensive debate on the biochemical intricacies of vitamin B metabolic pathways and how their deficiencies may affect the development of CKD, diabetes, and subsequently DKD, and vice-versa. Our article provides a review of updated evidence on the biochemical and physiological properties of the vitamin B sub-forms in normal states, and how vitamin B deficiency and defects in their metabolic pathways may influence CKD/DKD pathophysiology, and in reverse how CKD/DKD progression may affect vitamin B metabolism. We hope our article increases awareness of vitamin B deficiency in DKD and the complex physiological associations that exist between vitamin B deficiency, diabetes, and CKD. Further research efforts are needed going forward to address the knowledge gaps on this topic.
Collapse
Affiliation(s)
- Henry H L Wu
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, The University of Sydney, Sydney, NSW 2065, Australia
| | - Thomas McDonnell
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - Rajkumar Chinnadurai
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M1 7HR, UK
| |
Collapse
|
3
|
Kong Y, Liu P, Li Y, Nolan ND, Quinn PMJ, Hsu C, Jenny LA, Zhao J, Cui X, Chang Y, Wert KJ, Sparrow JR, Wang N, Tsang SH. HIF2α activation and mitochondrial deficit due to iron chelation cause retinal atrophy. EMBO Mol Med 2023; 15:e16525. [PMID: 36645044 PMCID: PMC9906391 DOI: 10.15252/emmm.202216525] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Iron accumulation causes cell death and disrupts tissue functions, which necessitates chelation therapy to reduce iron overload. However, clinical utilization of deferoxamine (DFO), an iron chelator, has been documented to give rise to systemic adverse effects, including ocular toxicity. This study provided the pathogenic and molecular basis for DFO-related retinopathy and identified retinal pigment epithelium (RPE) as the target tissue in DFO-related retinopathy. Our modeling demonstrated the susceptibility of RPE to DFO compared with the neuroretina. Intriguingly, we established upregulation of hypoxia inducible factor (HIF) 2α and mitochondrial deficit as the most prominent pathogenesis underlying the RPE atrophy. Moreover, suppressing hyperactivity of HIF2α and preserving mitochondrial dysfunction by α-ketoglutarate (AKG) protects the RPE against lesions both in vitro and in vivo. This supported our observation that AKG supplementation alleviates visual impairment in a patient undergoing DFO-chelation therapy. Overall, our study established a significant role of iron deficiency in initiating DFO-related RPE atrophy. Inhibiting HIF2α and rescuing mitochondrial function by AKG protect RPE cells and can potentially ameliorate patients' visual function.
Collapse
Affiliation(s)
- Yang Kong
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Pei‐Kang Liu
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
- Department of OphthalmologyKaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiungTaiwan
- School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Institute of Biomedical SciencesNational Sun Yat‐sen UniversityKaohsiungTaiwan
| | - Yao Li
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Nicholas D Nolan
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
- Department of Biomedical Engineering, The Fu Foundation School of Engineering and Applied ScienceColumbia UniversityNew YorkNYUSA
| | - Peter M J Quinn
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Chun‐Wei Hsu
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Laura A Jenny
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Jin Zhao
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Xuan Cui
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Ya‐Ju Chang
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Katherine J Wert
- Departments of Ophthalmology and Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- The Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Janet R Sparrow
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Nan‐Kai Wang
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| | - Stephen H Tsang
- Department of Ophthalmology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNYUSA
| |
Collapse
|
4
|
Lu S, Chen Y, Wang S, Han B, Zhao C, Xue P, Zhang Y, Fang H, Wang B, Cao Y. Combined metabolomic and transcriptomic analysis reveals key components of OsCIPK17 overexpression improves drought tolerance in rice. FRONTIERS IN PLANT SCIENCE 2023; 13:1043757. [PMID: 36699859 PMCID: PMC9868928 DOI: 10.3389/fpls.2022.1043757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Oryza Sativa is one of the most important food crops in China, which is easily affected by drought during its growth and development. As a member of the calcium signaling pathway, CBL-interacting protein kinase (CIPK) plays an important role in plant growth and development as well as environmental stress. However, there is no report on the function and mechanism of OsCIPK17 in rice drought resistance. We combined transcriptional and metabonomic analysis to clarify the specific mechanism of OsCIPK17 in response to rice drought tolerance. The results showed that OsCIPK17 improved drought resistance of rice by regulating deep roots under drought stress; Response to drought by regulating the energy metabolism pathway and controlling the accumulation of citric acid in the tricarboxylic acid (TCA) cycle; Our exogenous experiments also proved that OsCIPK17 responds to citric acid, and this process involves the auxin metabolism pathway; Exogenous citric acid can improve the drought resistance of overexpression plants. Our research reveals that OsCIPK17 positively regulates rice drought resistance and participates in the accumulation of citric acid in the TCA cycle, providing new insights for rice drought resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Baohua Wang
- *Correspondence: Baohua Wang, ; Yunying Cao,
| | - Yunying Cao
- *Correspondence: Baohua Wang, ; Yunying Cao,
| |
Collapse
|
5
|
Chen G, Hong X, He W, Ou L, Chen B, Zhong W, Lin Y, Luo X. The construction and analysis of tricarboxylic acid cycle related prognostic model for cervical cancer. Front Genet 2023; 14:1092276. [PMID: 36968582 PMCID: PMC10033772 DOI: 10.3389/fgene.2023.1092276] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/08/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction: Cervical cancer (CC) is the fourth most common malignant tumor in term of in incidence and mortality among women worldwide. The tricarboxylic acid (TCA) cycle is an important hub of energy metabolism, networking one-carbon metabolism, fatty acyl metabolism and glycolysis. It can be seen that the reprogramming of cell metabolism including TCA cycle plays an indispensable role in tumorigenesis and development. We aimed to identify genes related to the TCA cycle as prognostic markers in CC. Methods: Firstly, we performed the differential expressed analysis the gene expression profiles associated with TCA cycle obtained from The Cancer Genome Atlas (TCGA) database. Differential gene list was generated and cluster analysis was performed using genes with detected fold changes >1.5. Based on the subclusters of CC, we analysed the relationship between different clusters and clinical information. Next, Cox univariate and multivariate regression analysis were used to screen genes with prognostic characteristics, and risk scores were calculated according to the genes with prognostic characteristics. Additionally, we analyzed the correlation between the predictive signature and the treatment response of CC patients. Finally, we detected the expression of ench prognostic gene in clinical CC samples by quantitative polymerase chain reaction (RT-qPCR). Results: We constructed a prognostic model consist of seven TCA cycle associated gene (ACSL1, ALDOA, FOXK2, GPI, MDH1B, MDH2, and MTHFD1). Patients with CC were separated into two groups according to median risk score, and high-risk group had a worse prognosis compared to the low-risk group. High risk group had lower level of sensitivity to the conventional chemotherapy drugs including cisplatin, paclitaxel, sunitinib and docetaxel. The expression of ench prognostic signature in clinical CC samples was verified by qRT-PCR. Conclusion: There are several differentially expressed genes (DEGs) related to TCA cycle in CC. The risk score model based on these genes can effectively predict the prognosis of patients and provide tumor markers for predicting the prognosis of CC.
Collapse
Affiliation(s)
- Guanqiao Chen
- Guangzhou Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Women and Children Medical Hospital, Guangzhou, China
| | - Xiaoshan Hong
- Department of Gynecology, Guangdong Women and Children Medical Hospital, Guangzhou, China
| | - Wanshan He
- Guangzhou Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Women and Children Medical Hospital, Guangzhou, China
| | - Lingling Ou
- Guangzhou Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Women and Children Medical Hospital, Guangzhou, China
| | - Bin Chen
- Guangzhou Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Women and Children Medical Hospital, Guangzhou, China
| | - Weitao Zhong
- Department of Surgical Neonatal Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Yu Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Yu Lin, ; Xiping Luo,
| | - Xiping Luo
- Guangzhou Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Women and Children Medical Hospital, Guangzhou, China
- *Correspondence: Yu Lin, ; Xiping Luo,
| |
Collapse
|
6
|
Liu K, Du Y, Li H, Lin X. Identification of super-enhancer-associated transcription factors regulating glucose metabolism in poorly differentiated thyroid carcinoma. Genet Mol Biol 2022; 45:e20210370. [PMID: 36121916 PMCID: PMC9495016 DOI: 10.1590/1678-4685-gmb-2021-0370] [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: 11/23/2021] [Accepted: 06/05/2022] [Indexed: 12/24/2022] Open
Abstract
This study aimed to uncover transcription factors that regulate super-enhancers involved in glucose metabolism reprogramming in poorly differentiated thyroid carcinoma (PDTC). TCA cycle and pyruvate metabolism were significantly enriched in PDTC. Differentially expressed genes in PDTC vs. normal control tissues were located in key steps in TCA cycle and pyruvate metabolism. A total of 23 upregulated genes localized in TCA cycle and pyruvate metabolism were identified as super-enhancer-controlled genes. Transcription factor analysis of these 23 super-enhancer-controlled genes related to glucose metabolism was performed, and 20 transcription factors were obtained, of which KLF12, ZNF281 and RELA had a significant prognostic impact. Regulatory network of KLF12, ZNF281 and RELA controlled the expression of these four prognostic target genes (LDHA, ACLY, ME2 and IDH2). In vitro validation showed that silencing of KLF12, ZNF281 and RELA suppressed proliferation, glucose uptake, lactate production and ATP level, but increased ADP/ATP ratio in PDTC cells. In conclusion, KLF12, ZNF281 and RELA were identified as the key transcription factors that regulate super-enhancer-controlled genes related to glucose metabolism in PDTC. Our findings contribute to a deeper understanding of the regulatory mechanisms associated with glucose metabolism in PDTC, and advance the theoretical development of PDTC-targeted therapies.
Collapse
Affiliation(s)
- Kun Liu
- Tianjin Hospital, Endocrinology Department, Tianjin, P. R. China
| | - Yongrui Du
- 80th Group Military Hospital, Chinese Peoples Liberation Army, Endocrinology Department, Weifang, Shandong, P. R. China
| | - Hui Li
- XingTai Medical College, Basic Experiment Center, Xingtai, Hebei, P. R. China
| | - Xuexia Lin
- XingTai Medical College, Basic Experiment Center, Xingtai, Hebei, P. R. China
| |
Collapse
|
7
|
Han Y, Quan X, Chuang Y, Liang Q, Li Y, Yuan Z, Bian Y, Wei L, Wang J, Zhao Y. A multi-omics analysis for the prediction of neurocognitive disorders risk among the elderly in Macao. Clin Transl Med 2022; 12:e909. [PMID: 35696554 PMCID: PMC9191869 DOI: 10.1002/ctm2.909] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Due to the increasing ageing population, neurocognitive disorders (NCDs) have been a global public health issue, and its prevention and early diagnosis are crucial. Our previous study demonstrated that there is a significant correlation between specific populations and NCDs, but the biological characteristics of the vulnerable group predispose to NCDs are unclear. The purpose of this study is to investigate the predictors for the vulnerable group by a multi-omics analysis. METHODS Multi-omics approaches, including metagenomics, metabolomic and proteomic, were used to detect gut microbiota, faecal metabolites and urine exosome of 8 normal controls and 13 vulnerable elders after a rigorous screening of 400 elders in Macao. The multi-omics data were analysed using R and Bioconductor. The two-sided Wilcoxon's rank-sum test, Kruskal-Wallis rank sum test and the linear discriminant analysis effective size were applied to investigate characterized features. Moreover, a 2-year follow-up was conducted to evaluate cognitive function change of the elderly. RESULTS Compared with the control elders, the metagenomics of gut microbiota showed that Ruminococcus gnavus, Lachnospira eligens, Escherichia coli and Desulfovibrio piger were increased significantly in the vulnerable group. Carboxylates, like alpha-ketoglutaric acid and d-saccharic acid, and levels of vitamins had obvious differences in the faecal metabolites. There was a distinct decrease in the expression of eukaryotic translation initiation factor 2 subunit 1 (eIF2α) and amine oxidase A (MAO-A) according to the proteomic results of the urine exosomes. Moreover, the compound annual growth rate of neurocognitive scores was notably decreased in vulnerable elders. CONCLUSIONS The multi-omics characteristics of disturbed glyoxylate and dicarboxylate metabolism (bacteria), vitamin digestion and absorption and tricarboxylic acid cycle in vulnerable elders can serve as predictors of NCDs risk among the elderly of Macao. Intervention with them may be effective therapeutic approaches for NCDs, and the underlying mechanisms merit further exploration.
Collapse
Affiliation(s)
- Yan Han
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Xingping Quan
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | | | - Qiaoxing Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Yang Li
- Department of Gastrointestinal SurgerySecond Clinical Medical College of Jinan University, Shenzhen People's HospitalShenzhenChina
| | - Zhen Yuan
- Centre for Cognitive and Brain SciencesUniversity of MacauTaipaMacao SARChina
| | - Ying Bian
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Ji Wang
- School of Traditional Chinese MedicineBeijing University of Chinese MedicineBeijingChina
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| |
Collapse
|
8
|
Tancheva L, Lazarova M, Velkova L, Dolashki A, Uzunova D, Minchev B, Petkova-Kirova P, Hassanova Y, Gavrilova P, Tasheva K, Taseva T, Hodzhev Y, Atanasov AG, Stefanova M, Alexandrova A, Tzvetanova E, Atanasov V, Kalfin R, Dolashka P. Beneficial Effects of Snail Helix aspersa Extract in an Experimental Model of Alzheimer’s Type Dementia. J Alzheimers Dis 2022; 88:155-175. [DOI: 10.3233/jad-215693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background: Alzheimer’s disease (AD) is a complex neurodegenerative disease with multifactorial etiology, unsatisfactory treatment, and a necessity for broad-spectrum active substances for cure. The mucus from Helix aspersa snail is a mixture of bioactive molecules with antimicrobial, anti-inflammatory, antioxidant, and anti-apoptotic effects. So far there are no data concerning the capacity of snail extract (SE) to affect neurodegenerative disorders. Objective: The effects of SE from Helix aspersa on learning and memory deficits in Alzheimer’s type dementia (ATD) induced by scopolamine (Sco) in male Wistar rats were examined and some mechanisms of action underlying these effects were evaluated. Methods: SE (0.5 mL/100 g) was applied orally through a food tube for 16 consecutive days: 5 days before and 11 days simultaneously with Sco (2 mg/kg, intraperitoneally). At the end of Sco treatment, using behavioral methods, we evaluated memory performance. Additionally, in cortex and hippocampus the acetylcholinesterase (AChE) activity, acetylcholine and monoamines (dopamine, noradrenaline, and serotonin) content, levels of main oxidative stress markers, and expression of brain-derived neurotrophic factor (BDNF) and cAMP response element-binding protein (CREB) were determined. Results: We demonstrated that, according to all behavioral tests used, SE significantly improved the cognitive deficits induced by Sco. Furthermore, SE possessed AChE inhibitory activity, moderate antioxidant properties and the ability to modulate monoamines content in two brain structures. Moreover, multiple SE applications not only restored the depressed by Sco expression of CREB and BDNF, but significantly upregulated it. Conclusion: Summarizing results, we conclude that complex mechanisms underlie the beneficial effects of SE on impaired memory in Alzheimer’s type dementia.
Collapse
Affiliation(s)
- Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
- Weston Professor of Weizmann Institute of Science, Israel
| | - Maria Lazarova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Lyudmila Velkova
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Alexander Dolashki
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Borislav Minchev
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | | | - Yozljam Hassanova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Petja Gavrilova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Teodora Taseva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yordan Hodzhev
- National Center for Infectious and Parasitic Diseases, Sofia, Bulgaria
| | - Atanas G. Atanasov
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Magdalenka, Poland
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | | | - Albena Alexandrova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Elina Tzvetanova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Ventseslav Atanasov
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
- Department of Healthcare, South-West University “Neofit Rilski”, Blagoevgrad, Bulgaria
| | - Pavlina Dolashka
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
9
|
Mathew D, Luo Q, Bhatwadekar AD. Circadian Rhythm Disruption Results in Visual Dysfunction. FASEB Bioadv 2022; 4:364-378. [PMID: 35664832 PMCID: PMC9164246 DOI: 10.1096/fba.2021-00125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 11/14/2022] Open
Abstract
Artificial light has been increasingly in use for the past 70 years. The aberrant light exposure and round‐the‐clock nature of work lead to the disruption of biological clock. Circadian rhythm disruption (CRD) contributes to multiple metabolic and neurodegenerative diseases. However, its effect on vision is not understood. Moreover, the mammalian retina possesses an autonomous clock that could be reset with light exposure. We evaluated the impact of CRD on retinal morphology, physiology, and vision after housing mice in a disruption inducing shorter light/dark cycle (L10:D10). Interestingly, the mice under L10:D10 exhibited three different entrainment behaviors; “entrained,” “free‐running,” and “zigzagging.” These behavior groups under CRD exhibited reduced visual acuity, retinal thinning, and a decrease in the number of photoreceptors. Intriguingly, the electroretinogram response was decreased only in the mice exhibiting “entrained” behavior. The retinal proteome showed distinct changes with each entrainment behavior, and there was a dysfunctional oxidative stress‐antioxidant mechanism. These results demonstrate that CRD alters entrainment behavior and leads to visual dysfunction in mice. Our studies uniquely show the effect of entrainment behavior on retinal physiology. Our data have broader implications in understanding and mitigating the impact of CRD on vision and its potential role in the etiology of retinal diseases.
Collapse
Affiliation(s)
- Deepa Mathew
- Department of Ophthalmology Indiana University Indianapolis IN USA
| | - Qianyi Luo
- Department of Ophthalmology Indiana University Indianapolis IN USA
| | | |
Collapse
|
10
|
de Oliveira J, Farias HR, Streck EL. Experimental evidence of tyrosine neurotoxicity: focus on mitochondrial dysfunction. Metab Brain Dis 2021; 36:1673-1685. [PMID: 34212298 DOI: 10.1007/s11011-021-00781-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/07/2021] [Indexed: 11/29/2022]
Abstract
Tissue exposure to high levels of tyrosine, which is characteristic of an inborn error of metabolism named Tyrosinemia, is related to severe symptoms, including neurological alterations. The clinical manifestations and pathogenesis of tyrosine neurotoxicity can be recapitulated in experimental models in vivo and in vitro. A widely used experimental model to study brain tyrosine damage is the chronic and acute administration of this amino acid in infant rats. Other research groups and we have extensively studied the pathogenic events in the brain structures of rats exposed to high tyrosine levels. Rats administered acutely and chronically with tyrosine presented decreased and inhibition of the essential metabolism enzymes, e.g., Krebs cycle enzymes and mitochondrial respiratory complexes in the brain structures. These alterations induced by tyrosine toxicity were associated with brain oxidative stress, astrocytes, and, ultimately, cognitive impairments. Notably, in vivo data were corroborated by in vitro studies using cerebral regions homogenates incubated with tyrosine excess. Considering metabolism's importance to brain functioning, we hypothesized that mitochondrial and metabolic dysfunctions are closely related to neurological alterations induced by tyrosine neurotoxicity. Herein, we reviewed the main mechanisms associated with tyrosine neurotoxicity in experimental models, emphasizing the role of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jade de Oliveira
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-000, Brazil
| | - Hémelin Resende Farias
- Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-000, Brazil
| | - Emilio Luiz Streck
- Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, 88806-000, Brazil.
- Laboratório de Doenças Neurometabólicas, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, Brazil.
| |
Collapse
|
11
|
Yu Q, Pan H, Shao H, Qian C, Han J, Li Y, Lou Y. UPLC/MS-based untargeted metabolomics reveals the changes in muscle metabolism of electron beam irradiated Solenocera melantho during refrigerated storage. Food Chem 2021; 367:130713. [PMID: 34359006 DOI: 10.1016/j.foodchem.2021.130713] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 11/04/2022]
Abstract
Shrimp meat is an extremely perishable product; however, refrigeration can slow down spoilage. In this study, we used electron beam irradiation (EBI) to pre-treat shrimp meat and analyzed the metabolites of the treated shrimp meat during refrigerated storage using metabonomic analysis methods. In total, 4865 metabolites were identified, of which, 103 differential metabolites had KEGG (Kyoto Encyclopedia of Genes and Genomes) IDs. Further, two potential biomarkers were obtained. Based on the results, l-lysine was downregulated, while 2'-deoxyguanosine 5'-monophosphate and dihydroxyacetone phosphate acyl ester were upregulated during the refrigerated storage. The metabolic activity began to weaken gradually after 9 days. However, the different metabolites related to EBI were not identified herein. Nonetheless, the study findings revealed the metabolic changes in Solenocera melantho at the molecular level during refrigerated storage after EBI.
Collapse
Affiliation(s)
- Qi Yu
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Huijuan Pan
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Haitao Shao
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Chenru Qian
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Jiajun Han
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China; Sinopec Zhenhai Refining & Chemical Company, Ningbo, Zhejiang 315207, PR China
| | - Yongyong Li
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China.
| | - Yongjiang Lou
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China.
| |
Collapse
|
12
|
Kortesniemi M, Slupsky CM, Aatsinki AK, Sinkkonen J, Karlsson L, Linderborg KM, Yang B, Karlsson H, Kailanto HM. Human milk metabolome is associated with symptoms of maternal psychological distress and milk cortisol. Food Chem 2021; 356:129628. [PMID: 33836356 DOI: 10.1016/j.foodchem.2021.129628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/05/2021] [Accepted: 03/13/2021] [Indexed: 12/18/2022]
Abstract
The composition of human milk is subject to considerable variation, but the effects of maternal stress are largely unknown. We studied differences in human milk metabolome between Finnish mothers (n = 120, secretors) with symptoms of prenatal symptoms of psychological distress and milk cortisol concentrations. Human milk samples acquired at 2.5 months postpartum were analyzed using targeted 1H NMR metabolomics. Self-reported scores for depression (EPDS), overall anxiety (SCL-90), and pregnancy-related anxiety (PRAQ) were used to evaluate psychological distress. Prenatal psychological distress was positively associated with concentrations of short-chain fatty acids, caprate, and hypoxanthine (q < 0.0012). Milk cortisol was positively associated with lactate concentration (q < 0.05). Changes in the human milk metabolome were shown to be associated with maternal psychological distress and concentration of milk cortisol in a dissimilarly, suggesting alterations in bacterial and energy metabolism of the mother, respectively.
Collapse
Affiliation(s)
- Maaria Kortesniemi
- Food Chemistry and Food Development, Department of Life Technologies, University of Turku, Finland; Department of Nutrition, University of California Davis, Davis, CA, USA.
| | - Carolyn M Slupsky
- Department of Nutrition, University of California Davis, Davis, CA, USA; Department of Food Science and Technology, University of California Davis, Davis, CA, USA
| | - Anna-Katariina Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| | - Jari Sinkkonen
- Instrument Centre, Department of Chemistry, University of Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland; Department of Psychiatry, Turku University Hospital and University of Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Finland
| | - Kaisa M Linderborg
- Food Chemistry and Food Development, Department of Life Technologies, University of Turku, Finland
| | - Baoru Yang
- Food Chemistry and Food Development, Department of Life Technologies, University of Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland; Department of Psychiatry, Turku University Hospital and University of Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Finland
| | - Henna-Maria Kailanto
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Finland
| |
Collapse
|
13
|
Gasmi A, Peana M, Arshad M, Butnariu M, Menzel A, Bjørklund G. Krebs cycle: activators, inhibitors and their roles in the modulation of carcinogenesis. Arch Toxicol 2021; 95:1161-1178. [DOI: 10.1007/s00204-021-02974-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
|
14
|
Cai Z, Li CF, Han F, Liu C, Zhang A, Hsu CC, Peng D, Zhang X, Jin G, Rezaeian AH, Wang G, Zhang W, Pan BS, Wang CY, Wang YH, Wu SY, Yang SC, Hsu FC, D'Agostino RB, Furdui CM, Kucera GL, Parks JS, Chilton FH, Huang CY, Tsai FJ, Pasche B, Watabe K, Lin HK. Phosphorylation of PDHA by AMPK Drives TCA Cycle to Promote Cancer Metastasis. Mol Cell 2020; 80:263-278.e7. [PMID: 33022274 PMCID: PMC7534735 DOI: 10.1016/j.molcel.2020.09.018] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 01/28/2023]
Abstract
Cancer metastasis accounts for the major cause of cancer-related deaths. How disseminated cancer cells cope with hostile microenvironments in secondary site for full-blown metastasis is largely unknown. Here, we show that AMPK (AMP-activated protein kinase), activated in mouse metastasis models, drives pyruvate dehydrogenase complex (PDHc) activation to maintain TCA cycle (tricarboxylic acid cycle) and promotes cancer metastasis by adapting cancer cells to metabolic and oxidative stresses. This AMPK-PDHc axis is activated in advanced breast cancer and predicts poor metastasis-free survival. Mechanistically, AMPK localizes in the mitochondrial matrix and phosphorylates the catalytic alpha subunit of PDHc (PDHA) on two residues S295 and S314, which activates the enzymatic activity of PDHc and alleviates an inhibitory phosphorylation by PDHKs, respectively. Importantly, these phosphorylation events mediate PDHc function in cancer metastasis. Our study reveals that AMPK-mediated PDHA phosphorylation drives PDHc activation and TCA cycle to empower cancer cells adaptation to metastatic microenvironments for metastasis.
Collapse
Affiliation(s)
- Zhen Cai
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chien-Feng Li
- Department of Pathology, Chi-Mei Medical Center, Tainan 710, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan; Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Fei Han
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunfang Liu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anmei Zhang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Che-Chia Hsu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Danni Peng
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Xian Zhang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guoxiang Jin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abdol-Hossein Rezaeian
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guihua Wang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Weina Zhang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chi-Yun Wang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; International PhD Program in Innovative Technology of Biomedical Engineering and Medical Device, Ming Chi University of Technology, New Taipei City 243303, Taiwan
| | - Yu-Hui Wang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Shun-Chin Yang
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Fang-Chi Hsu
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Ralph B D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Christina M Furdui
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Gregory L Kucera
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John S Parks
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Floyd H Chilton
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Fuu-Jen Tsai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Boris Pasche
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
15
|
Wahl D, Anderson RM, Le Couteur DG. Antiaging Therapies, Cognitive Impairment, and Dementia. J Gerontol A Biol Sci Med Sci 2020; 75:1643-1652. [PMID: 31125402 PMCID: PMC7749193 DOI: 10.1093/gerona/glz135] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Indexed: 01/17/2023] Open
Abstract
Aging is a powerful risk factor for the development of many chronic diseases including dementia. Research based on disease models of dementia have yet to yield effective treatments, therefore it is opportune to consider whether the aging process itself might be a potential therapeutic target for the treatment and prevention of dementia. Numerous cellular and molecular pathways have been implicated in the aging process and compounds that target these processes are being developed to slow aging and delay the onset of age-associated conditions. A few particularly promising therapeutic agents have been shown to influence many of the main hallmarks of aging and increase life span in rodents. Here we discuss the evidence that some of these antiaging compounds may beneficially affect brain aging and thereby lower the risk for dementia.
Collapse
Affiliation(s)
- Devin Wahl
- Charles Perkins Centre
- Aging and Alzheimers Institute, ANZAC Research Institute, Centre for Education and Research on Ageing, The University of Sydney, Australia
| | - Rozalyn M Anderson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin
- Geriatrics Research Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - David G Le Couteur
- Charles Perkins Centre
- Aging and Alzheimers Institute, ANZAC Research Institute, Centre for Education and Research on Ageing, The University of Sydney, Australia
| |
Collapse
|
16
|
Vassilev NG, Simova SD, Dangalov M, Velkova L, Atanasov V, Dolashki A, Dolashka P. An 1H NMR- and MS-Based Study of Metabolites Profiling of Garden Snail Helix aspersa Mucus. Metabolites 2020; 10:E360. [PMID: 32887291 PMCID: PMC7570211 DOI: 10.3390/metabo10090360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/30/2022] Open
Abstract
Metabolic profiling based on 1H nuclear magnetic resonance (NMR) spectroscopy was applied with the aim to investigate the functional role of the metabolites in lyophilized mucus from the garden snail Helix aspersa. Twenty metabolites were unambiguously identified by 1H, 1D TOCSY, 2D J-resolved, 2D COSY, and 2D HSQC NMR spectra with water suppression. The metabolic profiles of two fractions with low molecular weight (Mw < 1 kDa and Mw < 3 kDa) are very similar. Metabolites with known antioxidant, antibacterial, and antimicrobial activity were detected by NMR metabolic analysis of mucus samples from Helix aspersa. Some of them were confirmed by mass spectrometric analysis. The primary structure of several peptides was identified in low molecular weight fractions (Mw < 1 kDa) by tandem mass spectrometry.
Collapse
Affiliation(s)
- Nikolay G. Vassilev
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 9, Acad. G. Bonchev Str., 1113 Sofia, Bulgaria; (S.D.S.); (M.D.); (L.V.); (V.A.); (A.D.)
| | | | | | | | | | | | - Pavlinka Dolashka
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 9, Acad. G. Bonchev Str., 1113 Sofia, Bulgaria; (S.D.S.); (M.D.); (L.V.); (V.A.); (A.D.)
| |
Collapse
|
17
|
Intermittent Fasting Enhanced the Cognitive Function in Older Adults with Mild Cognitive Impairment by Inducing Biochemical and Metabolic changes: A 3-Year Progressive Study. Nutrients 2020; 12:nu12092644. [PMID: 32872655 PMCID: PMC7551340 DOI: 10.3390/nu12092644] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/15/2022] Open
Abstract
Intermittent fasting (IF) refers to various dietary regimens that cycle between a period of non-fasting and a period of total fasting. This study aimed to determine the effects of IF on cognitive function among elderly individuals who practice IF who have mild cognitive impairment (MCI). A total of 99 elderly subjects with MCI of Malay ethnicity without any terminal illness were recruited from a larger cohort study, LRGS TUA. The subjects were divided into three groups, comprising those who were regularly practicing IF (r-IF), irregularly practicing IF (i-IF), and non-fasters (n-IF). Upon 36 months of follow-up, more MCI subjects in the r-IF group reverted to successful aging with no cognitive impairment and diseases (24.3%) compared to those in i-IF (14.2%) and n-IF groups (3.7%). The r-IF group’s subjects exhibited significant increment in superoxide dismutase (SOD) activity and reduction in body weight, levels of insulin, fasting blood glucose, malondialdehyde (MDA), C-reactive protein (CRP), and DNA damage. Moreover, metabolomics analysis showed that IF may modulate cognitive function via various metabolite pathways, including the synthesis and degradation of ketone bodies, butanoate metabolism, pyruvate metabolism, and glycolysis and gluconeogenesis pathways. Overall, the MCI-afflicted older adults who practiced IF regularly had better cognitive scores and reverted to better cognitive function at 36 months follow-up.
Collapse
|
18
|
Calandra MJ, Wang Y. POV remediation agents: α‐ketoglutarate salts and the treatment of citrus oils and citrus‐based model fragrances. FLAVOUR FRAG J 2020. [DOI: 10.1002/ffj.3607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
19
|
Combined metabolome and transcriptome analysis reveals key components of complete desiccation tolerance in an anhydrobiotic insect. Proc Natl Acad Sci U S A 2020; 117:19209-19220. [PMID: 32723826 PMCID: PMC7431039 DOI: 10.1073/pnas.2003650117] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Anhydrobiosis is a reversible ametabolic state that occurs in response to severe desiccation. The largest anhydrobiotic animal known is the larva of the African chironomid Polypedilum vanderplanki. Here, we investigated how the metabolism of larvae changes during the desiccation–rehydration cycle and how simple biochemical processes determine viability of the chironomid. Major findings suggest that, in addition to its known anhydroprotectant role, trehalose acts as a major source of energy for rehydration. Citrate and adenosine monophosphate, accumulated in the dry state, allow rapid resumption of metabolism during the recovery phase. Finally, metabolic waste is stored as stable or nontoxic compounds such as allantoin, xanthurenic acid, or ophthalmic acid that may also act as antioxidants. Some organisms have evolved a survival strategy to withstand severe dehydration in an ametabolic state, called anhydrobiosis. The only known example of anhydrobiosis among insects is observed in larvae of the chironomid Polypedilum vanderplanki. Recent studies have led to a better understanding of the molecular mechanisms underlying anhydrobiosis and the action of specific protective proteins. However, gene regulation alone cannot explain the rapid biochemical reactions and independent metabolic changes that are expected to sustain anhydrobiosis. For this reason, we conducted a comprehensive comparative metabolome–transcriptome analysis in the larvae. We showed that anhydrobiotic larvae adopt a unique metabolic strategy to cope with complete desiccation and, in particular, to allow recovery after rehydration. We argue that trehalose, previously known for its anhydroprotective properties, plays additional vital roles, providing both the principal source of energy and also the restoration of antioxidant potential via the pentose phosphate pathway during the early stages of rehydration. Thus, larval viability might be directly dependent on the total amount of carbohydrate (glycogen and trehalose). Furthermore, in the anhydrobiotic state, energy is stored as accumulated citrate and adenosine monophosphate, allowing rapid reactivation of the citric acid cycle and mitochondrial activity immediately after rehydration, before glycolysis is fully functional. Other specific adaptations to desiccation include potential antioxidants (e.g., ophthalmic acid) and measures to avoid the accumulation of toxic waste metabolites by converting these to stable and inert counterparts (e.g., xanthurenic acid and allantoin). Finally, we confirmed that these metabolic adaptations correlate with unique organization and expression of the corresponding enzyme genes.
Collapse
|
20
|
Drosophila SLC22 Orthologs Related to OATs, OCTs, and OCTNs Regulate Development and Responsiveness to Oxidative Stress. Int J Mol Sci 2020; 21:ijms21062002. [PMID: 32183456 PMCID: PMC7139749 DOI: 10.3390/ijms21062002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022] Open
Abstract
The SLC22 family of transporters is widely expressed, evolutionarily conserved, and plays a major role in regulating homeostasis by transporting small organic molecules such as metabolites, signaling molecules, and antioxidants. Analysis of transporters in fruit flies provides a simple yet orthologous platform to study the endogenous function of drug transporters in vivo. Evolutionary analysis of Drosophila melanogaster putative SLC22 orthologs reveals that, while many of the 25 SLC22 fruit fly orthologs do not fall within previously established SLC22 subclades, at least four members appear orthologous to mammalian SLC22 members (SLC22A16:CG6356, SLC22A15:CG7458, CG7442 and SLC22A18:CG3168). We functionally evaluated the role of SLC22 transporters in Drosophila melanogaster by knocking down 14 of these genes. Three putative SLC22 ortholog knockdowns-CG3168, CG6356, and CG7442/SLC22A-did not undergo eclosion and were lethal at the pupa stage, indicating the developmental importance of these genes. Additionally, knocking down four SLC22 members increased resistance to oxidative stress via paraquat testing (CG4630: p < 0.05, CG6006: p < 0.05, CG6126: p < 0.01 and CG16727: p < 0.05). Consistent with recent evidence that SLC22 is central to a Remote Sensing and Signaling Network (RSSN) involved in signaling and metabolism, these phenotypes support a key role for SLC22 in handling reactive oxygen species.
Collapse
|
21
|
Chang CH, Liu ZZ, Lee TH. Changes in hypothermal stress-induced hepatic mitochondrial metabolic patterns between fresh water- and seawater-acclimated milkfish, Chanos chanos. Sci Rep 2019; 9:18502. [PMID: 31811227 PMCID: PMC6897891 DOI: 10.1038/s41598-019-55055-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/21/2019] [Indexed: 11/09/2022] Open
Abstract
Milkfish (Chanos chanos) is a tropical euryhaline species. It can acclimate to fresh water (FW) or seawater (SW) and be cultured in both. In winter, cold snaps cause huge losses in milkfish revenue. Compared to FW-acclimated individuals, SW-acclimated milkfish have better low-temperature tolerance. Under hypothermal stress, a stable energy supply is critical to maintain normal liver function. In this study, the levels of key mitochondrial enzymes (citrate synthase (CS) and cytochrome c oxidase (COX)) in milkfish livers were examined. The CS:COX activity ratio in FW milkfish significantly increased under hypothermal stress (18 °C) whereas ATP (the end product of aerobic metabolism) was downregulated. Therefore, the activities of the enzymes involved in mitochondrial amino acid biosynthesis (aspartate aminotransferase (AST) and glutamate dehydrogenase (GDH)) were evaluated to elucidate energy flow in milkfish livers under hypothermal stress. In FW milkfish, GDH activity was upregulated whereas AST activity was downregulated. Nevertheless, the levels of all the aforementioned enzymes did not significantly change in SW milkfish under hypothermal stress. In summary, we clarified the mechanism accounting for the fact that SW milkfish have superior low-temperature tolerance to FW milkfish and demonstrated that SW and FW milkfish have different and unique strategies for regulating energy flow.
Collapse
Affiliation(s)
- Chia-Hao Chang
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan.,iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan
| | - Zong-Zheng Liu
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan
| | - Tsung-Han Lee
- Department of Life Sciences, National Chung Hsing University, Taichung, 402, Taiwan. .,iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, 402, Taiwan.
| |
Collapse
|
22
|
Di Paolo ML, Cervelli M, Mariottini P, Leonetti A, Polticelli F, Rosini M, Milelli A, Basagni F, Venerando R, Agostinelli E, Minarini A. Exploring the activity of polyamine analogues on polyamine and spermine oxidase: methoctramine, a potent and selective inhibitor of polyamine oxidase. J Enzyme Inhib Med Chem 2019; 34:740-752. [PMID: 30829081 PMCID: PMC6407594 DOI: 10.1080/14756366.2019.1584620] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/06/2019] [Accepted: 02/14/2019] [Indexed: 01/16/2023] Open
Abstract
Fourteen polyamine analogues, asymmetric or symmetric substituted spermine (1-9) or methoctramine (10-14) analogues, were evaluated as potential inhibitors or substrates of two enzymes of the polyamine catabolic pathway, spermine oxidase (SMOX) and acetylpolyamine oxidase (PAOX). Compound 2 turned out to be the best substrate for PAOX, having the highest affinity and catalytic efficiency with respect to its physiological substrates. Methoctramine (10), a well-known muscarinic M2 receptor antagonist, emerged as the most potent competitive PAOX inhibitor known so far (Ki = 10 nM), endowed with very good selectivity compared with SMOX (Ki=1.2 μM vs SMOX). The efficacy of methoctramine in inhibiting PAOX activity was confirmed in the HT22 cell line. Methoctramine is a very promising tool in the design of drugs targeting the polyamine catabolism pathway, both to understand the physio-pathological role of PAOX vs SMOX and for pharmacological applications, being the polyamine pathway involved in various pathologies.
Collapse
Affiliation(s)
| | | | | | | | - Fabio Polticelli
- Department of Sciences, University of Roma Tre, Roma, Italy
- Roma Tre Section, National Institute of Nuclear Physics, Roma, Italy
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Andrea Milelli
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Rimini, Italy
| | - Filippo Basagni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Rina Venerando
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Enzo Agostinelli
- Department of Biochemical Science "A. Rossi Fanelli", University of Rome "La Sapienza", Rome, Italy
- International Polyamines Foundation – ONLUS –Via del Forte Tiburtino 98, Rome, Italy
| | - Anna Minarini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
23
|
Fountain JC, Yang L, Pandey MK, Bajaj P, Alexander D, Chen S, Kemerait RC, Varshney RK, Guo B. Carbohydrate, glutathione, and polyamine metabolism are central to Aspergillus flavus oxidative stress responses over time. BMC Microbiol 2019; 19:209. [PMID: 31488075 PMCID: PMC6727485 DOI: 10.1186/s12866-019-1580-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/25/2019] [Indexed: 01/08/2023] Open
Abstract
Background The primary and secondary metabolites of fungi are critical for adaptation to environmental stresses, host pathogenicity, competition with other microbes, and reproductive fitness. Drought-derived reactive oxygen species (ROS) have been shown to stimulate aflatoxin production and regulate in Aspergillus flavus, and may function in signaling with host plants. Here, we have performed global, untargeted metabolomics to better understand the role of aflatoxin production in oxidative stress responses, and also explore isolate-specific oxidative stress responses over time. Results Two field isolates of A. flavus, AF13 and NRRL3357, possessing high and moderate aflatoxin production, respectively, were cultured in medium with and without supplementation with 15 mM H2O2, and mycelia were collected following 4 and 7 days in culture for global metabolomics. Overall, 389 compounds were described in the analysis which encompassed 9 biological super-pathways and 47 sub-pathways. These metabolites were examined for differential accumulation. Significant differences were observed in both isolates in response to oxidative stress and when comparing sampling time points. Conclusions The moderately high aflatoxin-producing isolate, NRRL3357, showed extensive stimulation of antioxidant mechanisms and pathways including polyamines metabolism, glutathione metabolism, TCA cycle, and lipid metabolism while the highly aflatoxigenic isolate, AF13, showed a less vigorous response to stress. Carbohydrate pathway levels also imply that carbohydrate repression and starvation may influence metabolite accumulation at the later timepoint. Higher conidial oxidative stress tolerance and antioxidant capacity in AF13 compared to NRRL3357, inferred from their metabolomic profiles and growth curves over time, may be connected to aflatoxin production capability and aflatoxin-related antioxidant accumulation. The coincidence of several of the detected metabolites in H2O2-stressed A. flavus and drought-stressed hosts also suggests their potential role in the interaction between these organisms and their use as markers/targets to enhance host resistance through biomarker selection or genetic engineering. Electronic supplementary material The online version of this article (10.1186/s12866-019-1580-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jake C Fountain
- USDA-ARS, Crop Protection and Management Research Unit, Tifton, GA, 31793, USA.,Department of Plant Pathology, University of Georgia, Tifton, GA, 31793, USA
| | - Liming Yang
- Department of Plant Pathology, University of Georgia, Tifton, GA, 31793, USA.,College of Biology and Environmental Science, Nanjing Forestry University, Nanjing, 210037, China
| | - Manish K Pandey
- International Crop Research Institute for the Semi-Arid Tropics (ICRISAT), Hyderabad, Telangana, 502324, India
| | - Prasad Bajaj
- International Crop Research Institute for the Semi-Arid Tropics (ICRISAT), Hyderabad, Telangana, 502324, India
| | | | - Sixue Chen
- Department of Biology, Genetics Institute, and Plant Molecular & Cellular Biology Program, University of Florida, Gainesville, FL, 32611, USA
| | - Robert C Kemerait
- Department of Plant Pathology, University of Georgia, Tifton, GA, 31793, USA
| | - Rajeev K Varshney
- International Crop Research Institute for the Semi-Arid Tropics (ICRISAT), Hyderabad, Telangana, 502324, India
| | - Baozhu Guo
- USDA-ARS, Crop Protection and Management Research Unit, Tifton, GA, 31793, USA.
| |
Collapse
|
24
|
Shimizu K, Matsuoka Y. Redox rebalance against genetic perturbations and modulation of central carbon metabolism by the oxidative stress regulation. Biotechnol Adv 2019; 37:107441. [PMID: 31472206 DOI: 10.1016/j.biotechadv.2019.107441] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/04/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022]
Abstract
The micro-aerophilic organisms and aerobes as well as yeast and higher organisms have evolved to gain energy through respiration (via oxidative phosphorylation), thereby enabling them to grow much faster than anaerobes. However, during respiration, reactive oxygen species (ROSs) are inherently (inevitably) generated, and threaten the cell's survival. Therefore, living organisms (or cells) must furnish the potent defense systems to keep such ROSs at harmless level, where the cofactor balance plays crucial roles. Namely, NADH is the source of energy generation (catabolism) in the respiratory chain reactions, through which ROSs are generated, while NADPH plays important roles not only for the cell synthesis (anabolism) but also for detoxifying ROSs. Therefore, the cell must rebalance the redox ratio by modulating the fluxes of the central carbon metabolism (CCM) by regulating the multi-level regulation machinery upon genetic perturbations and the change in the growth conditions. Here, we discuss about how aerobes accomplish such cofactor homeostasis against redox perturbations. In particular, we consider how single-gene mutants (including pgi, pfk, zwf, gnd and pyk mutants) modulate their metabolisms in relation to cofactor rebalance (and also by adaptive laboratory evolution). We also discuss about how the overproduction of NADPH (by the pathway gene mutation) can be utilized for the efficient production of useful value-added chemicals such as medicinal compounds, polyhydroxyalkanoates, and amino acids, all of which require NADPH in their synthetic pathways. We then discuss about the metabolic responses against oxidative stress, where αketoacids play important roles not only for the coordination between catabolism and anabolism, but also for detoxifying ROSs by non-enzymatic reactions, as well as for reducing the production of ROSs by repressing the activities of the TCA cycle and respiration (via carbon catabolite repression). Thus, we discuss about the mechanisms (basic strategies) that modulate the metabolism from respiration to respiro-fermentative metabolism causing overflow, based on the role of Pyk activity, affecting the NADPH production at the oxidative pentose phosphate (PP) pathway, and the roles of αketoacids for the change in the source of energy generation from the oxidative phosphorylation to the substrate level phosphorylation.
Collapse
Affiliation(s)
- Kazuyuki Shimizu
- Kyushu institute of Technology, Iizuka, Fukuoka 820-8502, Japan; Institute of Advanced Biosciences, Keio university, Tsuruoka, Yamagata 997-0017, Japan.
| | - Yu Matsuoka
- Kyushu institute of Technology, Iizuka, Fukuoka 820-8502, Japan.
| |
Collapse
|
25
|
Carvalho-Silva M, Gomes LM, Gomes ML, Ferreira BK, Schuck PF, Ferreira GC, Dal-Pizzol F, de Oliveira J, Scaini G, Streck EL. Omega-3 fatty acid supplementation can prevent changes in mitochondrial energy metabolism and oxidative stress caused by chronic administration of L-tyrosine in the brain of rats. Metab Brain Dis 2019; 34:1207-1219. [PMID: 30949952 DOI: 10.1007/s11011-019-00411-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/25/2019] [Indexed: 12/13/2022]
Abstract
Deficiency of hepatic enzyme tyrosine aminotransferase characterizes the innate error of autosomal recessive disease Tyrosinemia Type II. Patients may develop neurological and developmental difficulties due to high levels of the amino acid tyrosine in the body. Mechanisms underlying the neurological dysfunction in patients are poorly known. Importantly, Tyrosinemia patients have deficient Omega-3 fatty acids (n-3 PUFA). Here, we investigated the possible neuroprotective effect of the treatment with n-3 PUFA in the alterations caused by chronic administration of L-tyrosine on important parameters of energetic metabolism and oxidative stress in the hippocampus, striatum and cerebral cortex of developing rats. Chronic administration of L-tyrosine causes a decrease in the citrate synthase (CS) activity in the hippocampus and cerebral cortex, as well as in the succinate dehydrogenase (SDH) and isocitrate dehydrogenase (IDH) activities, and an increase in the α-ketoglutarate dehydrogenase activity in the hippocampus. Moreover, in the striatum, L-tyrosine administration caused a decrease in the activities of CS, SDH, creatine kinase, and complexes I, II-III and IV of the mitochondrial respiratory chain. We also observed that the high levels of L-tyrosine are related to oxidative stress in the brain. Notably, supplementation of n-3 PUFA prevented the majority of the modifications caused by the chronic administration of L-tyrosine in the cerebral enzyme activities, as well as ameliorated the oxidative stress in the brain regions of rats. These results indicate a possible neuroprotective and antioxidant role for n-3 PUFA and may represent a new therapeutic approach and potential adjuvant therapy to Tyrosinemia Type II individuals.
Collapse
Affiliation(s)
- Milena Carvalho-Silva
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Lara M Gomes
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Maria L Gomes
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Bruna K Ferreira
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Patricia F Schuck
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Gustavo C Ferreira
- Laboratório de Neuroenergética e Erros Inatos do Metabolismo, Instituto de Biofísica Carlos Chagas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe Dal-Pizzol
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Jade de Oliveira
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
| | - Giselli Scaini
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Emilio L Streck
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, SC, Brazil.
- Laboratório de Neurologia Experimental, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil.
| |
Collapse
|
26
|
Rescue from galactose-induced death of Leigh Syndrome patient cells by pyruvate and NAD .. Cell Death Dis 2018; 9:1135. [PMID: 30429455 PMCID: PMC6235972 DOI: 10.1038/s41419-018-1179-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 01/08/2023]
Abstract
Cell models of mitochondrial complex I (CI) deficiency display activation of glycolysis to compensate for the loss in mitochondrial ATP production. This adaptation can mask other relevant deficiency-induced aberrations in cell physiology. Here we investigated the viability, mitochondrial morphofunction, ROS levels and ATP homeostasis of primary skin fibroblasts from Leigh Syndrome (LS) patients with isolated CI deficiency. These cell lines harbored mutations in nuclear DNA (nDNA)-encoded CI genes (NDUFS7, NDUFS8, NDUFV1) and, to prevent glycolysis upregulation, were cultured in a pyruvate-free medium in which glucose was replaced by galactose. Following optimization of the cell culture protocol, LS fibroblasts died in the galactose medium, whereas control cells did not. LS cell death was dose-dependently inhibited by pyruvate, malate, oxaloacetate, α-ketoglutarate, aspartate, and exogenous NAD+ (eNAD), but not by lactate, succinate, α-ketobutyrate, and uridine. Pyruvate and eNAD increased the cellular NAD+ content in galactose-treated LS cells to a different extent and co-incubation studies revealed that pyruvate-induced rescue was not primarily mediated by NAD+. Functionally, in LS cells glucose-by-galactose replacement increased mitochondrial fragmentation and mass, depolarized the mitochondrial membrane potential (Δψ), increased H2DCFDA-oxidizing ROS levels, increased mitochondrial ATP generation, and reduced the total cellular ATP content. These aberrations were differentially rescued by pyruvate and eNAD, supporting the conclusion that these compounds rescue galactose-induced LS cell death via different mechanisms. These findings establish a cell-based strategy for intervention testing and enhance our understanding of CI deficiency pathophysiology.
Collapse
|
27
|
Bhattacharjee A, Prasad SK, Banerjee O, Singh S, Banerjee A, Bose A, Pal S, Maji BK, Mukherjee S. Targeting mitochondria with folic acid and vitamin B 12 ameliorates nicotine mediated islet cell dysfunction. ENVIRONMENTAL TOXICOLOGY 2018; 33:988-1000. [PMID: 29972271 DOI: 10.1002/tox.22586] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/27/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
Nicotine, one of the well-known highly toxic components of cigarette smoke, causes a number of adverse health effects and diseases. Our previous study has shown that nicotine induces reactive oxygen species (ROS) in islet cell and disrupts islet cell mitochondrial membrane potential (ΔΨm). However, supplementation with folic acid and vitamin B12 were found effective against nicotine induced changes in pancreatic islet cells. But the toxicological effects and underlying mechanisms of nicotine-induced mitochondrial dysfunction is still unknown. In this study, nicotine exposure decreases mitochondrial enzymes (pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase, aconitase, malate dehydrogenase) activities by increasing cytosolic Ca2+ level which may contribute to increased mitochondrial ROS production by raising its flow to mitochondria. This in turn produces malondialdehyde and nitric oxide (NO) with a concomitant decrease in the activities of antioxidative enzymes and glutathione levels leading to loss of ΔΨm. Simultaneously, nicotine induces pancreatic islet cell apoptosis by modulating ΔΨm via increased cytosolic Ca2+ level, altered Bcl-2, Bax, cytochrome c, caspase-9, PARP expressions which were prevented by the supplementation of folic acid and vitamin B12 . In conclusion, nicotine alters islet cell mitochondrial redox status, apoptotic machinery, and enzymes to cause disruption in the ΔΨm and supplementation of folic acid and vitamin B12 possibly blunted all these mitochondrial alterations. Therefore, this study may help to determine the pathophysiology of nicotine-mediated islet cell mitochondrial dysfunction.
Collapse
Affiliation(s)
- Ankita Bhattacharjee
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| | - Shilpi Kumari Prasad
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| | - Oly Banerjee
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| | - Siddhartha Singh
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| | - Arnab Banerjee
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| | - Ananya Bose
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| | - Swagata Pal
- Department of Physiology, Raja Peary Mohan College, Uttarpara, Kotrung, Hooghly, West Bengal, India
| | - Bithin Kumar Maji
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| | - Sandip Mukherjee
- Department of Physiology, Serampore College, Serampore, Hooghly, West Bengal, India
| |
Collapse
|
28
|
Doran ML, Knee JM, Wang N, Rzezniczak TZ, Parkes TL, Li L, Merritt TJS. Metabolomic analysis of oxidative stress: Superoxide dismutase mutation and paraquat induced stress in Drosophila melanogaster. Free Radic Biol Med 2017; 113:323-334. [PMID: 29031835 DOI: 10.1016/j.freeradbiomed.2017.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/02/2017] [Accepted: 10/07/2017] [Indexed: 10/18/2022]
Abstract
Oxidative stress results in substantial biochemical and physiological perturbations in essentially all organisms. To determine the broad metabolic effects of oxidative stress, we have quantified the response in Drosophila melanogaster to both genetically and environmentally derived oxidative stress. Flies were challenged with loss of Superoxide dismutase activity or chronic or acute exposure to the oxidizing chemical paraquat. Metabolic changes were then quantified using a recently developed chemical isotope labeling (CIL) liquid chromatography - mass spectrometry (LC-MS) platform that targets the carboxylic acid and amine/phenol submetabolomes with high metabolic coverage. We discovered wide spread changes in both submetabolomes in response to all three types of stresses including: changes to the urea cycle, tryptophan metabolism, porphyrin metabolism, as well as a series of metabolic pathways involved in glutathione synthesis. Strikingly, while there are commonalities across the conditions, all three resulted in different metabolomic responses, with the greatest difference between the genetic and environmental responses. Genetic oxidative stress resulted in substantially more widespread effects, both in terms of the percent of the metabolome altered, and the magnitude of changes in individual metabolites. Chronic and acute environmental stress resulted in more similar responses although both were distinct from genetic stress. Overall, these results indicate that the metabolomic response to oxidative stress is complex, reaching across multiple metabolic pathways, with some shared features but with more features unique to different, specific stressors.
Collapse
Affiliation(s)
- Marney L Doran
- Department of Chemistry & Biochemistry, Laurentian University, Sudbury, ON, Canada P3E 2C6
| | - Jose M Knee
- Department of Chemistry & Biochemistry, Laurentian University, Sudbury, ON, Canada P3E 2C6
| | - Nan Wang
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Teresa Z Rzezniczak
- Department of Chemistry & Biochemistry, Laurentian University, Sudbury, ON, Canada P3E 2C6
| | - Tony L Parkes
- Faculty of Arts & Science - Biology, Nipissing University, North Bay, ON, Canada P1B 8L7
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Thomas J S Merritt
- Department of Chemistry & Biochemistry, Laurentian University, Sudbury, ON, Canada P3E 2C6.
| |
Collapse
|