1
|
Patel RR, Arun PP, Singh SK, Singh M. Mycobacterial biofilms: Understanding the genetic factors playing significant role in pathogenesis, resistance and diagnosis. Life Sci 2024; 351:122778. [PMID: 38879157 DOI: 10.1016/j.lfs.2024.122778] [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: 12/27/2023] [Revised: 05/25/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
Even though the genus Mycobacterium is a diverse group consisting of a majority of environmental bacteria known as non-tuberculous mycobacteria (NTM), it also contains some of the deadliest pathogens (Mycobacterium tuberculosis) in history associated with chronic disease called tuberculosis (TB). Formation of biofilm is one of the unique strategies employed by mycobacteria to enhance their ability to survive in hostile conditions. Biofilm formation by Mycobacterium species is an emerging area of research with significant implications for understanding its pathogenesis and treatment of related infections, specifically TB. This review provides an overview of the biofilm-forming abilities of different species of Mycobacterium and the genetic factors influencing biofilm formation with a detailed focus on M. tuberculosis. Biofilm-mediated resistance is a significant challenge as it can limit antibiotic penetration and promote the survival of dormant mycobacterial cells. Key genetic factors promoting biofilm formation have been explored such as the mmpL genes involved in lipid transport and cell wall integrity as well as the groEL gene essential for mature biofilm formation. Additionally, biofilm-mediated antibiotic resistance and pathogenesis highlighting the specific niches, sites of infection along with the possible mechanisms of biofilm dissemination have been discussed. Furthermore, drug targets within mycobacterial biofilm and their role as potential biomarkers in the development of rapid diagnostic tools have been highlighted. The review summarises the current understanding of the complex nature of Mycobacterium biofilm and its clinical implications, paving the way for advancements in the field of disease diagnosis, management and treatment against its multi-drug resistant species.
Collapse
Affiliation(s)
- Ritu Raj Patel
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Pandey Priya Arun
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Sudhir Kumar Singh
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Meenakshi Singh
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
2
|
Mailloux RJ. The emerging importance of the α-keto acid dehydrogenase complexes in serving as intracellular and intercellular signaling platforms for the regulation of metabolism. Redox Biol 2024; 72:103155. [PMID: 38615490 PMCID: PMC11021975 DOI: 10.1016/j.redox.2024.103155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024] Open
Abstract
The α-keto acid dehydrogenase complex (KDHc) class of mitochondrial enzymes is composed of four members: pyruvate dehydrogenase (PDHc), α-ketoglutarate dehydrogenase (KGDHc), branched-chain keto acid dehydrogenase (BCKDHc), and 2-oxoadipate dehydrogenase (OADHc). These enzyme complexes occupy critical metabolic intersections that connect monosaccharide, amino acid, and fatty acid metabolism to Krebs cycle flux and oxidative phosphorylation (OxPhos). This feature also imbues KDHc enzymes with the heightened capacity to serve as platforms for propagation of intracellular and intercellular signaling. KDHc enzymes serve as a source and sink for mitochondrial hydrogen peroxide (mtH2O2), a vital second messenger used to trigger oxidative eustress pathways. Notably, deactivation of KDHc enzymes through reversible oxidation by mtH2O2 and other electrophiles modulates the availability of several Krebs cycle intermediates and related metabolites which serve as powerful intracellular and intercellular messengers. The KDHc enzymes also play important roles in the modulation of mitochondrial metabolism and epigenetic programming in the nucleus through the provision of various acyl-CoAs, which are used to acylate proteinaceous lysine residues. Intriguingly, nucleosomal control by acylation is also achieved through PDHc and KGDHc localization to the nuclear lumen. In this review, I discuss emerging concepts in the signaling roles fulfilled by the KDHc complexes. I highlight their vital function in serving as mitochondrial redox sensors and how this function can be used by cells to regulate the availability of critical metabolites required in cell signaling. Coupled with this, I describe in detail how defects in KDHc function can cause disease states through the disruption of cell redox homeodynamics and the deregulation of metabolic signaling. Finally, I propose that the intracellular and intercellular signaling functions of the KDHc enzymes are controlled through the reversible redox modification of the vicinal lipoic acid thiols in the E2 subunit of the complexes.
Collapse
Affiliation(s)
- Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
3
|
Kobayashi H, Yoshimoto C, Matsubara S, Shigetomi H, Imanaka S. Altered Energy Metabolism, Mitochondrial Dysfunction, and Redox Imbalance Influencing Reproductive Performance in Granulosa Cells and Oocyte During Aging. Reprod Sci 2024; 31:906-916. [PMID: 37917297 DOI: 10.1007/s43032-023-01394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Female fertility decreases during aging. The development of effective therapeutic strategies to address the age-related decline in oocyte quality and quantity and its accurate diagnosis remain major challenges. In this review, we summarize our current understanding of the study of aging and infertility, focusing primarily on the molecular basis of energy metabolism, mitochondrial function, and redox homeostasis in granulosa cells and oocytes, and discuss perspectives on future research directions. Mitochondria serve as a central hub sensing a multitude of physiological processes, including energy production, cellular redox homeostasis, aging, and senescence. Young granulosa cells favor glycolysis and actively produce pyruvate, NADPH, and other metabolites. Oocytes rely on oxidative phosphorylation fueled by nutrients, metabolites, and antioxidants provided by the adjacent granulosa cells. A reduced cellular energy metabolism phenotype, including both aerobic glycolysis and mitochondrial respiration, is characteristic of older female granulosa cells compared with younger female granulosa cells. Aged oocytes become more susceptible to oxidative damage to cells and mitochondria because of further depletion of antioxidant-dependent ROS scavenging systems. Molecular perturbations of gene expression caused by a subtle change in the follicular fluid microenvironment adversely affect energy metabolism and mitochondrial dynamics in granulosa cells and oocytes, further causing redox imbalance and accelerating aging and senescence. Furthermore, recent advances in technology are beginning to identify biofluid molecular markers that may influence follicular development and oocyte quality. Accumulating evidence suggests that redox imbalance caused by abnormal energy metabolism and/or mitochondrial dysfunction is closely linked to the pathophysiology of age-related subfertility.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-Cho, Kashihara, 634-0813, Japan.
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan.
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, 2-897-5 Shichi-Jyonishi-Machi, Nara, 630-8581, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
- Department of Medicine, Kei Oushin Clinic, 5-2-6, Naruo-Cho, Nishinomiya, 663-8184, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, 3-3-17 Kitatomigaoka-Cho, Nara, 634-0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-Cho, Kashihara, 634-0813, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
| |
Collapse
|
4
|
Zhou T, Zhong Y, Zhang Y, Zhou Y. Pyruvate Dehydrogenase Complex in Neonatal Hypoxic-Ischemic Brain Injury. ACS Pharmacol Transl Sci 2024; 7:42-47. [PMID: 38230287 PMCID: PMC10789137 DOI: 10.1021/acsptsci.3c00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
The disruption of cerebral energy metabolism in relation to brain damage has been the subject of extensive research. However, the pyruvate dehydrogenase complex (PDHC), which is primarily characterized by poor cerebral energy metabolism following brain trauma, has received relatively little study in comparison to newborn hypoxic-ischemic brain injury. Mitochondrial PDHC, a multienzyme complex that functions as a crucial hub in energy metabolism and acts as a central metabolic node to mediate pyruvate oxidation after glycolysis and fuel the Krebs cycle to meet energy demands, has been reported to be one cause of energy metabolism dysfunction according to recent studies. Here we assess the potential mechanisms of neonatal hypoxic-ischemic brain injury-related brain dysfunction mediated by PDHC and further discuss the neuroprotective effects of therapeutic medicines that target PDHC activation. We also provide a summary of recent research on medicines that target PDHC in neonates with hypoxic-ischemic brain damage. Through an understanding of the mechanisms by which it is modulated and an investigation of the neuroprotective techniques available to activate brain PDHC and improve neonatal hypoxic-ischemic impairment, our review emphasizes the significance of PDHC impairment in neonatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Tao Zhou
- Department
of Pharmaceutical and Medical Equipment, Rongtong Bayi Orthopedic Hospital of China, Chengdu 610031, China
| | - Yuangao Zhong
- Department
of Pharmaceutical Preparation Rongtong Bayi Orthopedic Hospital Of
China, Chengdu 610031, China
| | - Yong Zhang
- Department
of Pharmaceutical Preparation Rongtong Bayi Orthopedic Hospital Of
China, Chengdu 610031, China
| | - Yue Zhou
- Department
of Pharmacy, Xindu District People’s
Hospital of Chengdu, Chengdu 610500, China
| |
Collapse
|
5
|
Abudurexiti S, Xu S, Sun Z, Jiang Y, Gong P. Glucose metabolic reprogramming-related parameters for the prediction of 28-day neurological prognosis and all-cause mortality in patients after cardiac arrest: a prospective single-center observational study. World J Emerg Med 2024; 15:197-205. [PMID: 38855365 PMCID: PMC11153361 DOI: 10.5847/wjem.j.1920-8642.2024.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/06/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND We aimed to observe the dynamic changes in glucose metabolic reprogramming-related parameters and their ability to predict neurological prognosis and all-cause mortality in cardiac arrest patients after the restoration of spontaneous circulation (ROSC). METHODS Adult cardiac arrest patients after ROSC who were admitted to the emergency or cardiac intensive care unit of the First Affiliated Hospital of Dalian Medical University from August 1, 2017, to May 30, 2021, were enrolled. According to 28-day survival, the patients were divided into a non-survival group (n=82) and a survival group (n=38). Healthy adult volunteers (n=40) of similar ages and sexes were selected as controls. The serum levels of glucose metabolic reprogramming-related parameters (lactate dehydrogenase [LDH], lactate and pyruvate), neuron-specific enolase (NSE) and interleukin 6 (IL-6) were measured on days 1, 3, and 7 after ROSC. The Acute Physiology and Chronic Health Evaluation II (APACHE II) score and Sequential Organ Failure Assessment (SOFA) score were calculated. The Cerebral Performance Category (CPC) score was recorded on day 28 after ROSC. RESULTS Following ROSC, the serum LDH (607.0 U/L vs. 286.5 U/L), lactate (5.0 mmol/L vs. 2.0 mmol/L), pyruvate (178.0 μmol/L vs. 70.9 μmol/L), and lactate/pyruvate ratio (34.1 vs. 22.1) significantly increased and were higher in the non-survivors than in the survivors on admission (all P<0.05). Moreover, the serum LDH, pyruvate, IL-6, APACHE II score, and SOFA score on days 1, 3 and 7 after ROSC were significantly associated with 28-day poor neurological prognosis and 28-day all-cause mortality (all P<0.05). The serum LDH concentration on day 1 after ROSC had an area under the receiver operating characteristic curve (AUC) of 0.904 [95% confidence interval [95% CI]: 0.851-0.957]) with 96.8% specificity for predicting 28-day neurological prognosis and an AUC of 0.950 (95% CI: 0.911-0.989) with 94.7% specificity for predicting 28-day all-cause mortality, which was the highest among the glucose metabolic reprogramming-related parameters tested. CONCLUSION Serum parameters related to glucose metabolic reprogramming were significantly increased after ROSC. Increased serum LDH and pyruvate levels, and lactate/pyruvate ratio may be associated with 28-day poor neurological prognosis and all-cause mortality after ROSC, and the predictive efficacy of LDH during the first week was superior to others.
Collapse
Affiliation(s)
- Subi Abudurexiti
- Department of Emergency Medicine, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Shihai Xu
- Department of Emergency Medicine, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Zhangping Sun
- Department of Emergency Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yi Jiang
- Department of Emergency Medicine, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Ping Gong
- Department of Emergency Medicine, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| |
Collapse
|
6
|
Zhang Y, Sun M, Zhao H, Wang Z, Shi Y, Dong J, Wang K, Wang X, Li X, Qi H, Zhao X. Neuroprotective Effects and Therapeutic Potential of Dichloroacetate: Targeting Metabolic Disorders in Nervous System Diseases. Int J Nanomedicine 2023; 18:7559-7581. [PMID: 38106446 PMCID: PMC10725694 DOI: 10.2147/ijn.s439728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 12/19/2023] Open
Abstract
Dichloroacetate (DCA) is an investigational drug used to treat lactic acidosis and malignant tumours. It works by inhibiting pyruvate dehydrogenase kinase and increasing the rate of glucose oxidation. Some studies have documented the neuroprotective benefits of DCA. By reviewing these studies, this paper shows that DCA has multiple pharmacological activities, including regulating metabolism, ameliorating oxidative stress, attenuating neuroinflammation, inhibiting apoptosis, decreasing autophagy, protecting the blood‒brain barrier, improving the function of endothelial progenitor cells, improving mitochondrial dynamics, and decreasing amyloid β-protein. In addition, DCA inhibits the enzyme that metabolizes it, which leads to peripheral neurotoxicity due to drug accumulation that may be solved by individualized drug delivery and nanovesicle delivery. In summary, in this review, we analyse the mechanisms of neuroprotection by DCA in different diseases and discuss the causes of and solutions to its adverse effects.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Meiyan Sun
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Hongxiang Zhao
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Zhengyan Wang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Yanan Shi
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Jianxin Dong
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Kaifang Wang
- Department of Anesthesia, Tangdu Hospital, Fourth Military Medical University, Xian, Shanxi Province, People’s Republic of China
| | - Xi Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Xingyue Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Haiyan Qi
- Department of Anesthesiology, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, People’s Republic of China
| | - Xiaoyong Zhao
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, People’s Republic of China
| |
Collapse
|
7
|
Guo Z, Zhang Y, Huang A, Ni Q, Zeng C. Phenylbutyrate and Dichloroacetate Enhance the Liquid-Stored Boar Sperm Quality via PDK1 and PDK3. Int J Mol Sci 2023; 24:17091. [PMID: 38069413 PMCID: PMC10707026 DOI: 10.3390/ijms242317091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Artificial insemination (AI) with liquid-stored semen is the most prevalent and efficient assisted reproduction technique in the modern pork industry. Pyruvate dehydrogenase complex component X (PDHX) was demonstrated to be associated with sperm metabolism and affected the boar sperm viability, motility, and fertility. Pyruvate Dehydrogenase Kinases (PDKs) are the key metabolic enzymes that regulate pyruvate dehydrogenase complex (PDHC) activity and also the conversion from glycolysis to oxidative phosphorylation. In the present study, two PDK inhibitors, Dichloroacetate (DCA) and Phenylbutyrate (4-PBA), were added to an extender and investigated to determine their regulatory roles in liquid-stored boar sperm at 17 °C. The results indicated that PDK1 and PDK3 were predominantly located at the head and flagella of the boar sperm. The addition of 2 mM DCA and 0.5 mM 4-PBA significantly enhanced the sperm motility, plasma membrane integrity (PMI), mitochondrial membrane potential (MMP), and ATP content. In addition, DCA and 4-PBA exerted their effects by inhibiting PDK1 and PDK3, respectively. In conclusion, DCA and 4-PBA were found to regulate the boar sperm metabolic activities via PDK1 and PDK3. These both can improve the quality parameters of liquid-stored boar sperm, which will help to improve and optimize liquid-stored boar semen after their addition in the extender.
Collapse
Affiliation(s)
- Zhihua Guo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| | - Yan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| | - Anqi Huang
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China;
| | - Qingyong Ni
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| | - Changjun Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China; (Z.G.); (Y.Z.); (Q.N.)
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611134, China
| |
Collapse
|
8
|
Lenahan A, Mietzsch U, Wood TR, Callahan KP, Weiss EM, Miller DE, German K, Natarajan N, Puia-Dumitrescu M, Esposito V, Kolnik S, Law JB. Characteristics, Genetic Testing, and Diagnoses of Infants with Neonatal Encephalopathy Not Due to Hypoxic Ischemic Encephalopathy: A Cohort Study. J Pediatr 2023; 260:113533. [PMID: 37269901 DOI: 10.1016/j.jpeds.2023.113533] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/29/2023] [Accepted: 05/30/2023] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To characterize the presentation and evaluation of infants with neonatal encephalopathy (NE) not due to hypoxic-ischemic encephalopathy (non-HIE NE) and to describe the genetic abnormalities identified. STUDY DESIGN Retrospective cohort study of 193 non-HIE NE neonates admitted to a level IV NICU from 2015 through 2019. For changes in testing over time, Cochrane-Armitage test for trend was used with a Bonferroni-corrected P-value, and comparison between groups was performed using Fisher exact test. RESULT The most common symptom of non-HIE NE was abnormal tone in 47% (90/193). Ten percent (19/193) died prior to discharge, and 48% of survivors (83/174) required medical equipment at discharge. Forty percent (77/193) underwent genetic testing as an inpatient. Of 52 chromosomal studies, 54 targeted tests, and 16 exome sequences, 10%, 41%, and 69% were diagnostic, respectively, with no difference in diagnostic rates between infants with and without an associated congenital anomaly and/or dysmorphic feature. Twenty-eight genetic diagnoses were identified. CONCLUSIONS Neonates with non-HIE NE have high rates of morbidity and mortality and may benefit from early genetic testing, even in the absence of other exam findings. This study broadens our knowledge of genetic conditions underlying non-HIE NE, which may enable families and care teams to anticipate the needs of the individual, allow early initiation of targeted therapies, and facilitate decisions surrounding goals of care.
Collapse
Affiliation(s)
- Arthur Lenahan
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Ulrike Mietzsch
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Thomas R Wood
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Katharine Press Callahan
- Department of Pediatrics, Children's Hospital of Philadelphia, PA; Department of Medical Ethics and Health Policy, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Elliott M Weiss
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Danny E Miller
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Kendell German
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Niranjana Natarajan
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA; Division of Pediatric Neurology, Department of Neurology, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Mihai Puia-Dumitrescu
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Valentine Esposito
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Sarah Kolnik
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA
| | - Janessa B Law
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital, Seattle, WA.
| |
Collapse
|
9
|
Hoiland RL, Robba C, Menon DK, Citerio G, Sandroni C, Sekhon MS. Clinical targeting of the cerebral oxygen cascade to improve brain oxygenation in patients with hypoxic-ischaemic brain injury after cardiac arrest. Intensive Care Med 2023; 49:1062-1078. [PMID: 37507572 PMCID: PMC10499700 DOI: 10.1007/s00134-023-07165-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
The cerebral oxygen cascade includes three key stages: (a) convective oxygen delivery representing the bulk flow of oxygen to the cerebral vascular bed; (b) diffusion of oxygen from the blood into brain tissue; and (c) cellular utilisation of oxygen for aerobic metabolism. All three stages may become dysfunctional after resuscitation from cardiac arrest and contribute to hypoxic-ischaemic brain injury (HIBI). Improving convective cerebral oxygen delivery by optimising cerebral blood flow has been widely investigated as a strategy to mitigate HIBI. However, clinical trials aimed at optimising convective oxygen delivery have yielded neutral results. Advances in the understanding of HIBI pathophysiology suggest that impairments in the stages of the oxygen cascade pertaining to oxygen diffusion and cellular utilisation of oxygen should also be considered in identifying therapeutic strategies for the clinical management of HIBI patients. Culprit mechanisms for these impairments may include a widening of the diffusion barrier due to peri-vascular oedema and mitochondrial dysfunction. An integrated approach encompassing both intra-parenchymal and non-invasive neuromonitoring techniques may aid in detecting pathophysiologic changes in the oxygen cascade and enable patient-specific management aimed at reducing the severity of HIBI.
Collapse
Affiliation(s)
- Ryan L Hoiland
- Division of Critical Care Medicine, Department of Medicine, Faculty of Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada.
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, Faculty of Health and Social Development, University of British Columbia Okanagan, Kelowna, BC, Canada.
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada.
- Collaborative Entity for REsearching Brain Ischemia (CEREBRI), University of British Columbia, Vancouver, BC, Canada.
| | - Chiara Robba
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - David K Menon
- Department of Medicine, University Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Claudio Sandroni
- Department of Intensive Care, Emergency Medicine and Anaesthesiology, Fondazione Policlinico Universitario "Agostino Gemelli", IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mypinder S Sekhon
- Division of Critical Care Medicine, Department of Medicine, Faculty of Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada
- Collaborative Entity for REsearching Brain Ischemia (CEREBRI), University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Woodward KE, Murthy P, Mineyko A, Mohammad K, Esser MJ. Identifying Genetic Susceptibility in Neonates With Hypoxic-Ischemic Encephalopathy: A Retrospective Case Series. J Child Neurol 2023; 38:16-24. [PMID: 36628482 DOI: 10.1177/08830738221147805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy is a clinical phenomenon that often results from perinatal asphyxia. To mitigate secondary neurologic injury, prompt initial assessment and diagnosis is needed to identify patients eligible for therapeutic hypothermia. However, occasionally neonates present with a clinical picture of hypoxic-ischemic encephalopathy without significant risk factors for perinatal asphyxia. We hypothesized that in patients with genetic abnormalities, the clinical manifestation of those abnormalities may overlap with hypoxic-ischemic encephalopathy criteria, potentially contributing to a causal misattribution. We reviewed 210 charts of infants meeting local protocol criteria for moderate to severe hypoxic-ischemic encephalopathy in neonatal intensive care units in Calgary, Alberta. All patients that met criteria for therapeutic hypothermia were eligible for the study. Data were collected surrounding pregnancy and birth histories, as well as any available genetic or metabolic testing including microarray, gene panels, whole-exome sequencing, and newborn metabolic screens. Twenty-eight patients had genetic testing such as microarray, whole-exome sequencing, or a gene panel, because of clinical suspicion. Ten of 28 patients had genetic mutations, including CDKL5, pyruvate dehydrogenase, CFTR, CYP21A2, ISY1, KIF1A, KCNQ2, SCN9A, MTFMT, and NPHP1. All patients lacked significant risk factors to support a moderate to severe hypoxic-ischemic encephalopathy diagnosis. Treatment was changed in 2 patients because of confirmed genetic etiology. This study demonstrates the importance of identifying genetic comorbidities as potential contributors to a hypoxic-ischemic encephalopathy phenotype in neonates. Early identification of clinical factors that support an alternate diagnosis should be considered when the patient's clinical picture is not typical of hypoxic-ischemic encephalopathy and could aid in both treatment decisions and outcome prognostication.
Collapse
Affiliation(s)
- Kristine E Woodward
- Department of Pediatrics, Section of Neurology, University of Calgary, Cumming School of Medicine, 9978Alberta Children's Hospital, Calgary, Canada.,Department of Neurosciences, University of Calgary, Cumming School of Medicine, 9978Alberta Children's Hospital, Calgary, Canada
| | - Prashanth Murthy
- Department of Pediatrics, Section of Neonatology, University of Calgary, Cumming School of Medicine, 9978Alberta Children's Hospital, Calgary, Canada
| | - Aleksandra Mineyko
- Department of Pediatrics, Section of Neurology, University of Calgary, Cumming School of Medicine, 9978Alberta Children's Hospital, Calgary, Canada
| | - Khorshid Mohammad
- Department of Pediatrics, Section of Neonatology, University of Calgary, Cumming School of Medicine, 9978Alberta Children's Hospital, Calgary, Canada
| | - Michael J Esser
- Department of Pediatrics, Section of Neurology, University of Calgary, Cumming School of Medicine, 9978Alberta Children's Hospital, Calgary, Canada.,Department of Neurosciences, University of Calgary, Cumming School of Medicine, 9978Alberta Children's Hospital, Calgary, Canada
| |
Collapse
|
11
|
Lee SH, Choi BY, Kho AR, Hong DK, Kang BS, Park MK, Lee SH, Choi HC, Song HK, Suh SW. Combined Treatment of Dichloroacetic Acid and Pyruvate Increased Neuronal Survival after Seizure. Nutrients 2022; 14:4804. [PMID: 36432491 PMCID: PMC9698956 DOI: 10.3390/nu14224804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
During seizure activity, glucose and Adenosine triphosphate (ATP) levels are significantly decreased in the brain, which is a contributing factor to seizure-induced neuronal death. Dichloroacetic acid (DCA) has been shown to prevent cell death. DCA is also known to be involved in adenosine triphosphate (ATP) production by activating pyruvate dehydrogenase (PDH), a gatekeeper of glucose oxidation, as a pyruvate dehydrogenase kinase (PDK) inhibitor. To confirm these findings, in this study, rats were given a per oral (P.O.) injection of DCA (100 mg/kg) with pyruvate (50 mg/kg) once per day for 1 week starting 2 h after the onset of seizures induced by pilocarpine administration. Neuronal death and oxidative stress were assessed 1 week after seizure to determine if the combined treatment of pyruvate and DCA increased neuronal survival and reduced oxidative damage in the hippocampus. We found that the combined treatment of pyruvate and DCA showed protective effects against seizure-associated hippocampal neuronal cell death compared to the vehicle-treated group. Treatment with combined pyruvate and DCA after seizure may have a therapeutic effect by increasing the proportion of pyruvate converted to ATP. Thus, the current research demonstrates that the combined treatment of pyruvate and DCA may have therapeutic potential in seizure-induced neuronal death.
Collapse
Affiliation(s)
- Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Bo Young Choi
- Department of Physical Education, Hallym University, Chuncheon 24252, Korea
- Institute of Sports Science, Hallym University, Chuncheon 24252, Korea
| | - A Ra Kho
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Si Hyun Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Hui Chul Choi
- College of Medicine, Neurology, Hallym University, Chuncheon 24252, Korea
- Hallym Institute of Epilepsy Research, Hallym University, Chuncheon 24252, Korea
| | - Hong Ki Song
- College of Medicine, Neurology, Hallym University, Chuncheon 24252, Korea
- Hallym Institute of Epilepsy Research, Hallym University, Chuncheon 24252, Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
- Hallym Institute of Epilepsy Research, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
12
|
Saha B, Sahu G, Sharma P. A Novel Therapeutic Approach With Sodium Pyruvate on Vital Signs, Acid–Base, and Metabolic Disturbances in Rats With a Combined Blast and Hemorrhagic Shock. Front Neurol 2022; 13:938076. [PMID: 36034304 PMCID: PMC9400716 DOI: 10.3389/fneur.2022.938076] [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: 05/06/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background Blast injuries from improvised explosive devices (IEDs) are known to cause blast traumatic brain injuries (bTBIs), hemorrhagic shock (HS), organ damage, mitochondrial dysfunction, and subsequent free radical production. A pre-citric acid cycle reagent, pyruvate, is suggested to improve mitochondrial ATP production through the activation of the mitochondrial gatekeeper enzyme “pyruvate dehydrogenase complex (PDH).” Our study aimed to investigate the role of physiologic, metabolic, and mitochondrial effects of hypertonic sodium pyruvate resuscitation in rats with a combined blast and HS injury. Methods A pre-clinical rat model of combined injury with repetitive 20 PSI blast exposure accompanied with HS and fluid resuscitation (sodium pyruvate as metabolic adjuvant or hypertonic saline as control), followed by transfusion of shed blood was used in this study. Control sham animals (instrumental and time-matched) received anesthesia and cannulation, but neither received any injury nor treatment. The mean arterial pressure and heart rate were recorded throughout the experiment by a computerized program. Blood collected at T0 (baseline), T60 (after HS), and T180 (end) was analyzed for blood chemistry and mitochondrial PDH enzyme activity. Results Sodium pyruvate resuscitation significantly improved the mean arterial pressure (MAP), heart rate (HR), pulse pressure (PP), hemodynamic stability (Shock index), and autonomic response (Kerdo index) after the HS and/or blast injury. Compared with the baseline values, plasma lactate and lactate/pyruvate ratios were significantly increased. In contrast, base excess BE/(HCO3-) was low and the pH was also acidotic <7.3, indicating the sign of metabolic acidosis after blast and HS in all animal groups. Sodium pyruvate infusion significantly corrected these parameters at the end of the experiment. The PDH activity also improved after the sodium pyruvate infusion. Conclusion In our rat model of a combined blast and HS injury, hypertonic sodium pyruvate resuscitation was significantly effective in hemodynamic stabilization by correcting the acid–base status and mitochondrial mechanisms via its pyruvate dehydrogenase enzyme.
Collapse
|
13
|
Li HL, Shan SW, Stamer WD, Li KK, Chan HHL, Civan MM, To CH, Lam TC, Do CW. Mechanistic Effects of Baicalein on Aqueous Humor Drainage and Intraocular Pressure. Int J Mol Sci 2022; 23:ijms23137372. [PMID: 35806375 PMCID: PMC9266486 DOI: 10.3390/ijms23137372] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Elevated intraocular pressure (IOP) is a major risk factor for glaucoma that results from impeded fluid drainage. The increase in outflow resistance is caused by trabecular meshwork (TM) cell dysfunction and excessive extracellular matrix (ECM) deposition. Baicalein (Ba) is a natural flavonoid and has been shown to regulate cell contraction, fluid secretion, and ECM remodeling in various cell types, suggesting the potential significance of regulating outflow resistance and IOP. We demonstrated that Ba significantly lowered the IOP by about 5 mmHg in living mice. Consistent with that, Ba increased the outflow facility by up to 90% in enucleated mouse eyes. The effects of Ba on cell volume regulation and contractility were examined in primary human TM (hTM) cells. We found that Ba (1–100 µM) had no effect on cell volume under iso-osmotic conditions but inhibited the regulatory volume decrease (RVD) by up to 70% under hypotonic challenge. In addition, Ba relaxed hTM cells via reduced myosin light chain (MLC) phosphorylation. Using iTRAQ-based quantitative proteomics, 47 proteins were significantly regulated in hTM cells after a 3-h Ba treatment. Ba significantly increased the expression of cathepsin B by 1.51-fold and downregulated the expression of D-dopachrome decarboxylase and pre-B-cell leukemia transcription factor-interacting protein 1 with a fold-change of 0.58 and 0.40, respectively. We suggest that a Ba-mediated increase in outflow facility is triggered by cell relaxation via MLC phosphorylation along with inhibiting RVD in hTM cells. The Ba-mediated changes in protein expression support the notion of altered ECM homeostasis, potentially contributing to a reduction of outflow resistance and thereby IOP.
Collapse
Affiliation(s)
- Hoi-lam Li
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong; (H.-l.L.); (S.W.S.); (K.-k.L.); (H.H.-l.C.); (C.-h.T.); (T.C.L.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Sze Wan Shan
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong; (H.-l.L.); (S.W.S.); (K.-k.L.); (H.H.-l.C.); (C.-h.T.); (T.C.L.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong
- Research Centre for Chinese Medicine Innovation (RCMI), The Hong Kong Polytechnic University, Hong Kong
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC 27708, USA;
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - King-kit Li
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong; (H.-l.L.); (S.W.S.); (K.-k.L.); (H.H.-l.C.); (C.-h.T.); (T.C.L.)
| | - Henry Ho-lung Chan
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong; (H.-l.L.); (S.W.S.); (K.-k.L.); (H.H.-l.C.); (C.-h.T.); (T.C.L.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong
- Research Centre for Chinese Medicine Innovation (RCMI), The Hong Kong Polytechnic University, Hong Kong
| | - Mortimer M. Civan
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Chi-ho To
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong; (H.-l.L.); (S.W.S.); (K.-k.L.); (H.H.-l.C.); (C.-h.T.); (T.C.L.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong
- Research Centre for Chinese Medicine Innovation (RCMI), The Hong Kong Polytechnic University, Hong Kong
| | - Thomas Chuen Lam
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong; (H.-l.L.); (S.W.S.); (K.-k.L.); (H.H.-l.C.); (C.-h.T.); (T.C.L.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong
- Research Centre for Chinese Medicine Innovation (RCMI), The Hong Kong Polytechnic University, Hong Kong
| | - Chi-wai Do
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong; (H.-l.L.); (S.W.S.); (K.-k.L.); (H.H.-l.C.); (C.-h.T.); (T.C.L.)
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong
- Research Centre for Chinese Medicine Innovation (RCMI), The Hong Kong Polytechnic University, Hong Kong
- Research Institute of Smart Ageing (RISA), The Hong Kong Polytechnic University, Hong Kong
- Correspondence:
| |
Collapse
|
14
|
Omori NE, Woo GH, Mansor LS. Exogenous Ketones and Lactate as a Potential Therapeutic Intervention for Brain Injury and Neurodegenerative Conditions. Front Hum Neurosci 2022; 16:846183. [PMID: 36267349 PMCID: PMC9577611 DOI: 10.3389/fnhum.2022.846183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic dysfunction is a ubiquitous underlying feature of many neurological conditions including acute traumatic brain injuries and chronic neurodegenerative conditions. A central problem in neurological patients, in particular those with traumatic brain injuries, is an impairment in the utilization of glucose, which is the predominant metabolic substrate in a normally functioning brain. In such patients, alternative substrates including ketone bodies and lactate become important metabolic candidates for maintaining brain function. While the potential neuroprotective benefits of ketosis have been recognized for up to almost a century, the majority of work has focused on the use of ketogenic diets to induce such a state, which is inappropriate in cases of acute disease due to the prolonged periods of time (i.e., weeks to months) required for the effects of a ketogenic diet to be seen. The following review seeks to explore the neuroprotective effects of exogenous ketone and lactate preparations, which have more recently become commercially available and are able to induce a deep ketogenic response in a fraction of the time. The rapid response of exogenous preparations makes their use as a therapeutic adjunct more feasible from a clinical perspective in both acute and chronic neurological conditions. Potentially, their ability to globally moderate long-term, occult brain dysfunction may also be relevant in reducing lifetime risks of certain neurodegenerative conditions. In particular, this review explores the association between traumatic brain injury and contusion-related dementia, assessing metabolic parallels and highlighting the potential role of exogenous ketone and lactate therapies.
Collapse
|
15
|
Wang L, Wu L, Fu Y, Jiang L, Huang Z, Yang Z, Fang X. Changes of Key Rate-Limiting Enzyme Activity in Glucose Metabolism After Cardiopulmonary Resuscitation. Shock 2022; 57:576-582. [PMID: 34731097 DOI: 10.1097/shk.0000000000001884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To investigate the activity of key rate-limiting enzymes of glucose metabolism after restoration of spontaneous circulation (ROSC), to explore the potential pathophysiological mechanism of impaired myocardial energy metabolism after cardiopulmonary resuscitation (CPR). METHODS Twenty-one male Sprague-Dawley rats were randomized into three experimental groups assigned in accordance with different observation times after ROSC: Sham, instrumented rats without induced cardiac arrest or resuscitation; post-resuscitation (PR2 h); PR24 h. In these groups, CPR, including precordial compressions and synchronized mechanical ventilation, was initiated 6 min after asphyxia-induced cardiac arrest. Hearts were harvested after ROSC and samples were used to detect high-energy phosphate and glucose metabolic enzyme activity. RESULTS Compared with sham, the contents of phosphocreatine and adenosine triphosphate reduced in the PR2 h group, while remained unchanged in the PR24 h group. Activities of hexokinase and pyruvate kinase did not change after ROSC. Phosphofructokinase activity decreased only in the PR24 h group. Activities of pyruvate dehydrogenase and citrate synthase fell in PR2 h group and recovered in the PR24 h group. However, isocitrate dehydrogenase and α-ketoglutarate dehydrogenase activities fell in the PR2 h group, but did not recover in the PR24 h group. CONCLUSIONS Lowered key rate-limiting enzymes activity in glucose metabolism resulted in impairment of energy production in the early stage of ROSC, but partially recovered in 24 h. This process has a role in the mechanism of impaired myocardial energy metabolism after CPR. This investigation might shed light on new strategies to treat post resuscitation myocardial dysfunction.
Collapse
Affiliation(s)
- Liwen Wang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liangliang Wu
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yue Fu
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Emergency Medicine, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Longyuan Jiang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zitong Huang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhengfei Yang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangshao Fang
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Katz A, Brosnahan SB, Papadopoulos J, Parnia S, Lam JQ. Pharmacologic neuroprotection in ischemic brain injury after cardiac arrest. Ann N Y Acad Sci 2021; 1507:49-59. [PMID: 34060087 DOI: 10.1111/nyas.14613] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/31/2022]
Abstract
Cardiac arrest has many implications for morbidity and mortality. Few interventions have been shown to improve return of spontaneous circulation (ROSC) and long-term outcomes after cardiac arrest. Ischemic-reperfusion injury upon achieving ROSC creates an imbalance between oxygen supply and demand. Multiple events occur in the postcardiac arrest period, including excitotoxicity, mitochondrial dysfunction, and oxidative stress and inflammation, all of which contribute to ongoing brain injury and cellular death. Given that complex pathophysiology underlies global brain hypoxic ischemia, neuroprotective strategies targeting multiple stages of the neuropathologic cascade should be considered as a means of mitigating secondary neuronal injury and improving neurologic outcomes and survival in cardiac arrest victims. In this review article, we discuss a number of different pharmacologic agents that may have a potential role in targeting these injurious pathways following cardiac arrest. Pharmacologic therapies most relevant for discussion currently include memantine, perampanel, magnesium, propofol, thiamine, methylene blue, vitamin C, vitamin E, coenzyme Q10 , minocycline, steroids, and aspirin.
Collapse
Affiliation(s)
- Alyson Katz
- Department of Pharmacy, NYU Langone Health, New York, New York
| | - Shari B Brosnahan
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University School of Medicine, New York, New York
| | | | - Sam Parnia
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University School of Medicine, New York, New York
| | - Jason Q Lam
- Division of Pulmonary and Critical Care, Department of Medicine, Kaiser Permanente South Sacramento Medical Center, Sacramento, California
| |
Collapse
|
17
|
Weiland J, Beez A, Westermaier T, Kunze E, Sirén AL, Lilla N. Neuroprotective Strategies in Aneurysmal Subarachnoid Hemorrhage (aSAH). Int J Mol Sci 2021; 22:5442. [PMID: 34064048 PMCID: PMC8196706 DOI: 10.3390/ijms22115442] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) remains a disease with high mortality and morbidity. Since treating vasospasm has not inevitably led to an improvement in outcome, the actual emphasis is on finding neuroprotective therapies in the early phase following aSAH to prevent secondary brain injury in the later phase of disease. Within the early phase, neuroinflammation, thromboinflammation, disturbances in brain metabolism and early neuroprotective therapies directed against delayed cerebral ischemia (DCI) came into focus. Herein, the role of neuroinflammation, thromboinflammation and metabolism in aSAH is depicted. Potential neuroprotective strategies regarding neuroinflammation target microglia activation, metalloproteases, autophagy and the pathway via Toll-like receptor 4 (TLR4), high mobility group box 1 (HMGB1), NF-κB and finally the release of cytokines like TNFα or IL-1. Following the link to thromboinflammation, potential neuroprotective therapies try to target microthrombus formation, platelets and platelet receptors as well as clot clearance and immune cell infiltration. Potential neuroprotective strategies regarding metabolism try to re-balance the mismatch of energy need and supply following aSAH, for example, in restoring fuel to the TCA cycle or bypassing distinct energy pathways. Overall, this review addresses current neuroprotective strategies in aSAH, hopefully leading to future translational therapy options to prevent secondary brain injury.
Collapse
Affiliation(s)
- Judith Weiland
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Alexandra Beez
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Thomas Westermaier
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
- Department of Neurosurgery, Helios-Amper Klinikum Dachau, Krankenhausstr. 15, 85221 Dachau, Germany
| | - Ekkehard Kunze
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
| | - Nadine Lilla
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany; (A.B.); (T.W.); (E.K.); (A.-L.S.)
- Department of Neurosurgery, University Hospital Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
18
|
Mi Y, Qi G, Brinton RD, Yin F. Mitochondria-Targeted Therapeutics for Alzheimer's Disease: The Good, the Bad, the Potential. Antioxid Redox Signal 2021; 34:611-630. [PMID: 32143551 PMCID: PMC7891225 DOI: 10.1089/ars.2020.8070] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022]
Abstract
Significance: Alzheimer's disease (AD) is the leading cause of dementia. Thus far, 99.6% of clinical trials, including those targeting energy metabolism, have failed to exert disease-modifying efficacy. Altered mitochondrial function and disruption to the brain bioenergetic system have long-been documented as early events during the pathological progression of AD. Recent Advances: While therapeutic approaches that directly promote mitochondrial bioenergetic machinery or eliminate reactive oxygen species have exhibited limited translatability, emerging strategies targeting nonenergetic aspects of mitochondria provide novel therapeutic targets with the potential to modify AD risk and progression. Growing evidence also reveals a critical link between mitochondrial phenotype and neuroinflammation via metabolic reprogramming of glial cells. Critical Issues: Herein, we summarize major classes of mitochondrion-centered AD therapeutic strategies. In addition, the discrepancy in their efficacy when translated from preclinical models to clinical trials is addressed. Key factors that differentiate the responsiveness to bioenergetic interventions, including sex, apolipoprotein E genotype, and cellular diversity in the brain, are discussed. Future Directions: We propose that the future development of mitochondria-targeted AD therapeutics should consider the interactions between bioenergetics and other disease mechanisms, which may require cell-type-specific targeting to distinguish neurons and non-neuronal cells. Moreover, a successful strategy will likely include stratification by metabolic phenotype, which varies by sex and genetic risk profile and dynamically changes throughout the course of disease. As the network of mitochondrial integration expands across intracellular and systems level biology, assessment of intended, the good, versus unintended consequences, the bad, will be required to reach the potential of mitochondrial therapeutics.
Collapse
Affiliation(s)
- Yashi Mi
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Guoyuan Qi
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
- Department of Pharmacology, College of Medicine Tucson, Tucson, Arizona, USA
- Department of Neurology, College of Medicine Tucson, Tucson, Arizona, USA
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, Arizona, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, Arizona, USA
- Department of Pharmacology, College of Medicine Tucson, Tucson, Arizona, USA
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
19
|
Calabrò M, Rinaldi C, Santoro G, Crisafulli C. The biological pathways of Alzheimer disease: a review. AIMS Neurosci 2020; 8:86-132. [PMID: 33490374 PMCID: PMC7815481 DOI: 10.3934/neuroscience.2021005] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer disease is a progressive neurodegenerative disorder, mainly affecting older people, which severely impairs patients' quality of life. In the recent years, the number of affected individuals has seen a rapid increase. It is estimated that up to 107 million subjects will be affected by 2050 worldwide. Research in this area has revealed a lot about the biological and environmental underpinnings of Alzheimer, especially its correlation with β-Amyloid and Tau related mechanics; however, the precise molecular events and biological pathways behind the disease are yet to be discovered. In this review, we focus our attention on the biological mechanics that may lie behind Alzheimer development. In particular, we briefly describe the genetic elements and discuss about specific biological processes potentially associated with the disease.
Collapse
Affiliation(s)
| | | | | | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| |
Collapse
|
20
|
Tanaka E, Ogawa Y, Fujii R, Shimonaka T, Sato Y, Hamazaki T, Nagamura-Inoue T, Shintaku H, Tsuji M. Metabolomic analysis and mass spectrometry imaging after neonatal stroke and cell therapies in mouse brains. Sci Rep 2020; 10:21881. [PMID: 33318553 PMCID: PMC7736587 DOI: 10.1038/s41598-020-78930-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic brain injury provokes complex, time-dependent downstream pathways that ultimately lead to cell death. We aimed to demonstrate the levels of a wide range of metabolites in brain lysates and their on-tissue distribution following neonatal stroke and cell therapies. Postnatal day 12 mice underwent middle cerebral artery occlusion (MCAO) and were administered 1 × 105 cells after 48 h. Metabolomic analysis of the injured hemisphere demonstrated that a variety of amino acids were significantly increased and that tricarboxylic acid cycle intermediates and some related amino acids, such as glutamate, were decreased. With the exception of the changes in citric acid, neither mesenchymal stem/stromal cells nor CD34+ cells ameliorated these changes. On-tissue visualization with matrix-assisted laser desorption/ionization-mass spectrometry (MALDI-MS) imaging revealed that the signal intensity of glutamate was significantly decreased in the infarct area, consistent with the metabolomic analysis, while its intensity was significantly increased in the peri-infarct area after MCAO. Although cell therapies did not ameliorate the changes in metabolites in the infarct area, mesenchymal stem cells ameliorated the increased levels of glutamate and carnitine in the peri-infarct area. MALDI-MS imaging showed the location-specific effect of cell therapies even in this subacute setting after MCAO. These methodologies may be useful for further investigation of possible treatments for ischemic brain injury.
Collapse
Affiliation(s)
- Emi Tanaka
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuko Ogawa
- Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Ritsuko Fujii
- Division of Bioenergetics, Research Center for Artificial Photosynthesis, Osaka City University, Osaka, Japan.,Division of Molecular Materials Science, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Tomomi Shimonaka
- Analysis Division, Research Center for Artificial Photosynthesis, Osaka City University, Osaka, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan. .,Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan.
| |
Collapse
|
21
|
Cura AJ, Xu X, Egan S, Aron K, Jenkins L, Hageman T, Huang Y, Chollangi S, Borys M, Ghose S, Li ZJ. Metabolic understanding of disulfide reduction during monoclonal antibody production. Appl Microbiol Biotechnol 2020; 104:9655-9669. [PMID: 32997205 DOI: 10.1007/s00253-020-10916-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 01/04/2023]
Abstract
The disulfide reduction of intact monoclonal antibodies (mAbs) and subsequent formation of low molecular weight (LMW) species pose a direct risk to product stability, potency, and patient safety. Although enzymatic mechanisms of reduction are well established, an understanding of the cellular mechanisms during the bioreactor process leading to increased risk of disulfide reduction after harvest remains elusive. In this study, we examined bench, pilot, and manufacturing-scale batches of two mAbs expressed in Chinese hamster ovary (CHO) cells, where harvested cell culture fluid (HCCF) occasionally demonstrated disulfide reduction. Comparative proteomics highlighted a significant elevation in glyceraldehyde-3-phosphate dehydrogenase (GAPDH) levels in a highly reducing batch of HCCF, compared to a non-reducing batch. Analysis during production cell culture showed that increased GAPDH gene and protein expression correlated to disulfide reduction risk in HCCF in every case examined. Additionally, glucose 6-phosphate dehydrogenase (G6PD) activity and an increased (≥ 300%) lactate/pyruvate molar ratio (lac/pyr) during production cell culture correlated to disulfide reduction risk, suggesting a metabolic shift to the pentose phosphate pathway (PPP). In all, these results suggest that metabolic alterations during cell culture lead to changes in protein expression and enzyme activity that in turn increase the risk of disulfide reduction in HCCF. KEY POINTS: • Bioreactor conditions resulted in reduction susceptible harvest material. • GAPDH expression, G6PD activity, and lac/pyr ratio correlated with mAb reduction. • Demonstrated role for cell metabolic changes in post-harvest mAb reduction. Graphical abstract.
Collapse
Affiliation(s)
- Anthony J Cura
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA
| | - Xuankuo Xu
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA.
| | - Susan Egan
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA
| | - Kathryn Aron
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA.
| | - Lauren Jenkins
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA
| | - Tyler Hageman
- Biophysical Characterization, Global Product Development and Supply, Bristol-Myers Squibb Company, 1 Squibb Drive, New Brunswick, NJ, 08901, USA
| | - Yunping Huang
- Biophysical Characterization, Global Product Development and Supply, Bristol-Myers Squibb Company, 1 Squibb Drive, New Brunswick, NJ, 08901, USA
| | - Srinivas Chollangi
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA
| | - Michael Borys
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA
| | - Sanchayita Ghose
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA
| | - Zheng Jian Li
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb Company, 38 Jackson Road, Devens, MA, 01434, USA
| |
Collapse
|
22
|
Chen S, Teng T, Wen S, Zhang T, Huang H. The aceE involves in mycolic acid synthesis and biofilm formation in Mycobacterium smegmatis. BMC Microbiol 2020; 20:259. [PMID: 32811434 PMCID: PMC7437000 DOI: 10.1186/s12866-020-01940-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/11/2020] [Indexed: 11/26/2022] Open
Abstract
Background The integrity of cell wall structure is highly significant for the in vivo survival of mycobacteria. We hypothesized that changes in morphology may indicate changes in cell wall metabolism and identified an aceE gene mutant (aceE-mut) which presented a deficient colony morphology on 7H10 agar by screening transposon mutagenesis in Mycolicibacterium smegmatis, basonym Mycobacterium smegmatis (M. smegmatis). This study aimed to identify the functional role of aceE gene in cell wall biosynthesis in M. smegmatis. Results We observed that the colony morphology of aceE-mut was quite different, smaller and smoother on the solid culture medium than the wild-type (WT) strain during the transposon library screening of M. smegmatis. Notably, in contrast with the WT, which aggregates and forms biofilm, the aceE-mut lost its ability of growing aggregately and biofilm formation, which are two very important features of mycobacteria. The morphological changes in the aceE-mut strain were further confirmed by electron microscopy which indicated smoother and thinner cell envelope images in contrast with the rough morphology of WT strains. Additionally, the aceE-mut was more fragile to acidic stress and exhibited a pronounced defects in entering the macrophages as compared to the WT. The analysis of mycolic acid (MA) using LC-MS indicated deficiency of alpha-MA and epoxy-MA in aceE-mut strain whereas complementation of the aceE-mut with a wild-type aceE gene restored the composition of MA. Conclusions Over all, this study indicates that aceE gene plays a significant role in the mycolic acid synthesis and affects the colony morphology, biofilm formation of M. smegmatis and bacteria invasion of macrophage.
Collapse
Affiliation(s)
- Suting Chen
- National Clinical Laboratory on Tuberculosis, Beijing Key laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Tianlu Teng
- National Clinical Laboratory on Tuberculosis, Beijing Key laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Shuan Wen
- National Clinical Laboratory on Tuberculosis, Beijing Key laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Tingting Zhang
- National Clinical Laboratory on Tuberculosis, Beijing Key laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Key laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, 101149, China.
| |
Collapse
|
23
|
Garabadu D, Agrawal N, Sharma A, Sharma S. Mitochondrial metabolism: a common link between neuroinflammation and neurodegeneration. Behav Pharmacol 2020; 30:642-652. [PMID: 31625975 DOI: 10.1097/fbp.0000000000000505] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neurodegenerative disorders have been considered as a growing health concern for decades. Increasing risk of neurodegenerative disorders creates a socioeconomic burden to both patients and care givers. Mitochondria are organelle that are involved in both neuroinflammation and neurodegeneration. There are few reports on the effect of mitochondrial metabolism on the progress of neurodegeneration and neuroinflammation. Therefore, the present review summarizes the potential contribution of mitochondrial metabolic pathways in the pathogenesis of neuroinflammation and neurodegeneration. Mitochondrial pyruvate metabolism plays a critical role in the pathogenesis of neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. However, there its potential contribution in other neurodegenerative disorders is as yet unproven. The mitochondrial pyruvate carrier and pyruvate dehydrogenase can modulate mitochondrial pyruvate metabolism to attenuate neuroinflammation and neurodegeneration. Further, it has been observed that the mitochondrial citric acid cycle can regulate the pathogenesis of neuroinflammation and neurodegeneration. Additional research should be undertaken to target tricarboxylic acid cycle enzymes to minimize the progress of neuroinflammation and neurodegeneration. It has also been observed that the mitochondrial urea cycle can potentially contribute to the progression of neurodegenerative disorders. Therefore, targeting this pathway may control the mitochondrial dysfunction-induced neuroinflammation and neurodegeneration. Furthermore, the mitochondrial malate-aspartate shuttle could be another target to control mitochondrial dysfunction-induced neuroinflammation and neurodegeneration in neurodegenerative disorders.
Collapse
Affiliation(s)
- Debapriya Garabadu
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, India
| | | | | | | |
Collapse
|
24
|
Marquez AM, Morgan RW, Ko T, Landis WP, Hefti MM, Mavroudis CD, McManus MJ, Karlsson M, Starr J, Roberts AL, Lin Y, Nadkarni V, Licht DJ, Berg RA, Sutton RM, Kilbaugh TJ. Oxygen Exposure During Cardiopulmonary Resuscitation Is Associated With Cerebral Oxidative Injury in a Randomized, Blinded, Controlled, Preclinical Trial. J Am Heart Assoc 2020; 9:e015032. [PMID: 32321350 PMCID: PMC7428577 DOI: 10.1161/jaha.119.015032] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Hyperoxia during cardiopulmonary resuscitation (CPR) may lead to oxidative injury from mitochondrial‐derived reactive oxygen species, despite guidelines recommending 1.0 inspired oxygen during CPR. We hypothesized exposure to 1.0 inspired oxygen during CPR would result in cerebral hyperoxia, higher mitochondrial‐derived reactive oxygen species, increased oxidative injury, and similar survival compared with those exposed to 21% oxygen. Methods and Results Four‐week‐old piglets (n=25) underwent asphyxial cardiac arrest followed by randomization and blinding to CPR with 0.21 (n=10) or 1.0 inspired oxygen (n=10) through 10 minutes post return of spontaneous circulation. Sham was n=5. Survivors received 4 hours of protocolized postarrest care, whereupon brain was obtained for mitochondrial analysis and neuropathology. Groups were compared using Kruskal‐Wallis test, Wilcoxon rank‐sum test, and generalized estimating equations regression models. Both 1.0 and 0.21 groups were similar in systemic hemodynamics and cerebral blood flow, as well as survival (8/10). The 1.0 animals had relative cerebral hyperoxia during CPR and immediately following return of spontaneous circulation (brain tissue oxygen tension, 85% [interquartile range, 72%–120%] baseline in 0.21 animals versus 697% [interquartile range, 515%–721%] baseline in 1.0 animals; P=0.001 at 10 minutes postarrest). Cerebral mitochondrial reactive oxygen species production was higher in animals treated with 1.0 compared with 0.21 (P<0.03). Exposure to 1.0 oxygen led to increased cerebral oxidative injury to proteins and lipids, as evidenced by significantly higher protein carbonyls and 4‐hydroxynoneals compared with 0.21 (P<0.05) and sham (P<0.001). Conclusions Exposure to 1.0 inspired oxygen during CPR caused cerebral hyperoxia during resuscitation, and resultant increased mitochondrial‐derived reactive oxygen species and oxidative injury following cardiac arrest.
Collapse
Affiliation(s)
- Alexandra M Marquez
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Ryan W Morgan
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Tiffany Ko
- Division of Neurology Department of Pediatrics Children's Hospital of Philadelphia PA
| | - William P Landis
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Marco M Hefti
- Department of Pathology University of Iowa Iowa City IA
| | - Constantine D Mavroudis
- Division of Cardiothoracic Surgery Department of Surgery Children's Hospital of Philadelphia PA
| | - Meagan J McManus
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | | | - Jonathan Starr
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Anna L Roberts
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Yuxi Lin
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Vinay Nadkarni
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Daniel J Licht
- Division of Neurology Department of Pediatrics Children's Hospital of Philadelphia PA
| | - Robert A Berg
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Robert M Sutton
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| | - Todd J Kilbaugh
- Division of Critical Care Medicine Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia PA
| |
Collapse
|
25
|
Hayashida K, Bagchi A, Miyazaki Y, Hirai S, Seth D, Silverman MG, Rezoagli E, Marutani E, Mori N, Magliocca A, Liu X, Berra L, Hindle AG, Donnino MW, Malhotra R, Bradley MO, Stamler JS, Ichinose F. Improvement in Outcomes After Cardiac Arrest and Resuscitation by Inhibition of S-Nitrosoglutathione Reductase. Circulation 2019; 139:815-827. [PMID: 30586713 DOI: 10.1161/circulationaha.117.032488] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The biological effects of nitric oxide are mediated via protein S-nitrosylation. Levels of S-nitrosylated protein are controlled in part by the denitrosylase, S-nitrosoglutathione reductase (GSNOR). The objective of this study was to examine whether GSNOR inhibition improves outcomes after cardiac arrest and cardiopulmonary resuscitation (CA/CPR). METHODS Adult wild-type C57BL/6 and GSNOR-deleted (GSNOR-/-) mice were subjected to potassium chloride-induced CA and subsequently resuscitated. Fifteen minutes after a return of spontaneous circulation, wild-type mice were randomized to receive the GSNOR inhibitor, SPL-334.1, or normal saline as placebo. Mortality, neurological outcome, GSNOR activity, and levels of S-nitrosylated proteins were evaluated. Plasma GSNOR activity was measured in plasma samples obtained from post-CA patients, preoperative cardiac surgery patients, and healthy volunteers. RESULTS GSNOR activity was increased in plasma and multiple organs of mice, including brain in particular. Levels of protein S-nitrosylation were decreased in the brain 6 hours after CA/CPR. Administration of SPL-334.1 attenuated the increase in GSNOR activity in brain, heart, liver, spleen, and plasma, and restored S-nitrosylated protein levels in the brain. Inhibition of GSNOR attenuated ischemic brain injury and improved survival in wild-type mice after CA/CPR (81.8% in SPL-334.1 versus 36.4% in placebo; log rank P=0.031). Similarly, GSNOR deletion prevented the reduction in the number of S-nitrosylated proteins in the brain, mitigated brain injury, and improved neurological recovery and survival after CA/CPR. Both GSNOR inhibition and deletion attenuated CA/CPR-induced disruption of blood brain barrier. Post-CA patients had higher plasma GSNOR activity than did preoperative cardiac surgery patients or healthy volunteers ( P<0.0001). Plasma GSNOR activity was positively correlated with initial lactate levels in postarrest patients (Spearman correlation coefficient=0.48; P=0.045). CONCLUSIONS CA and CPR activated GSNOR and reduced the number of S-nitrosylated proteins in the brain. Pharmacological inhibition or genetic deletion of GSNOR prevented ischemic brain injury and improved survival rates by restoring S-nitrosylated protein levels in the brain after CA/CPR in mice. Our observations suggest that GSNOR is a novel biomarker of postarrest brain injury as well as a molecular target to improve outcomes after CA.
Collapse
Affiliation(s)
- Kei Hayashida
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Aranya Bagchi
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Yusuke Miyazaki
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Shuichi Hirai
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Divya Seth
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center (D.S.), Cleveland, OH
| | - Michael G Silverman
- Cardiology Division, Department of Medicine, Massachusetts General Hospital (M.G.S., R.M.), Boston, MA
| | - Emanuele Rezoagli
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Eizo Marutani
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Naohiro Mori
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Aurora Magliocca
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Xiaowen Liu
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA (X.L., M.W.D.)
| | - Lorenzo Berra
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Allyson G Hindle
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Michael W Donnino
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA (X.L., M.W.D.)
| | - Rajeev Malhotra
- Cardiology Division, Department of Medicine, Massachusetts General Hospital (M.G.S., R.M.), Boston, MA
| | | | | | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| |
Collapse
|
26
|
The Effects of Sodium Dichloroacetate on Mitochondrial Dysfunction and Neuronal Death Following Hypoglycemia-Induced Injury. Cells 2019; 8:cells8050405. [PMID: 31052436 PMCID: PMC6562710 DOI: 10.3390/cells8050405] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/17/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022] Open
Abstract
Our previous studies demonstrated that some degree of neuronal death is caused by hypoglycemia, but a subsequent and more severe wave of neuronal cell death occurs due to glucose reperfusion, which results from the rapid restoration of low blood glucose levels. Mitochondrial dysfunction caused by hypoglycemia leads to increased levels of pyruvate dehydrogenase kinase (PDK) and suppresses the formation of ATP by inhibiting pyruvate dehydrogenase (PDH) activation, which can convert pyruvate into acetyl-coenzyme A (acetyl-CoA). Sodium dichloroacetate (DCA) is a PDK inhibitor and activates PDH, the gatekeeper of glucose oxidation. However, no studies about the effect of DCA on hypoglycemia have been published. In the present study, we hypothesized that DCA treatment could reduce neuronal death through improvement of glycolysis and prevention of reactive oxygen species production after hypoglycemia. To test this, we used an animal model of insulin-induced hypoglycemia and injected DCA (100 mg/kg, i.v., two days) following hypoglycemic insult. Histological evaluation was performed one week after hypoglycemia. DCA treatment reduced hypoglycemia-induced oxidative stress, microglial activation, blood–brain barrier disruption, and neuronal death compared to the vehicle-treated hypoglycemia group. Therefore, our findings suggest that DCA may have the therapeutic potential to reduce hippocampal neuronal death after hypoglycemia.
Collapse
|
27
|
Haque MM, Murale DP, Kim YK, Lee JS. Crosstalk between Oxidative Stress and Tauopathy. Int J Mol Sci 2019; 20:ijms20081959. [PMID: 31013607 PMCID: PMC6514575 DOI: 10.3390/ijms20081959] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/16/2019] [Accepted: 04/19/2019] [Indexed: 12/11/2022] Open
Abstract
Tauopathy is a collective term for neurodegenerative diseases associated with pathological modifications of tau protein. Tau modifications are mediated by many factors. Recently, reactive oxygen species (ROS) have attracted attention due to their upstream and downstream effects on tauopathy. In physiological conditions, healthy cells generate a moderate level of ROS for self-defense against foreign invaders. Imbalances between ROS and the anti-oxidation pathway cause an accumulation of excessive ROS. There is clear evidence that ROS directly promotes tau modifications in tauopathy. ROS is also highly upregulated in the patients’ brain of tauopathies, and anti-oxidants are currently prescribed as potential therapeutic agents for tauopathy. Thus, there is a clear connection between oxidative stress (OS) and tauopathies that needs to be studied in more detail. In this review, we will describe the chemical nature of ROS and their roles in tauopathy.
Collapse
Affiliation(s)
- Md Mamunul Haque
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.
| | - Dhiraj P Murale
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.
| | - Yun Kyung Kim
- Bio-Med Division, KIST-School UST, Seoul 02792, Korea.
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.
| | - Jun-Seok Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.
- Bio-Med Division, KIST-School UST, Seoul 02792, Korea.
| |
Collapse
|
28
|
Agoston DV, Kamnaksh A. Protein biomarkers of epileptogenicity after traumatic brain injury. Neurobiol Dis 2019; 123:59-68. [PMID: 30030023 PMCID: PMC6800147 DOI: 10.1016/j.nbd.2018.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a major risk factor for acquired epilepsy. Post-traumatic epilepsy (PTE) develops over time in up to 50% of patients with severe TBI. PTE is mostly unresponsive to traditional anti-seizure treatments suggesting distinct, injury-induced pathomechanisms in the development of this condition. Moderate and severe TBIs cause significant tissue damage, bleeding, neuron and glia death, as well as axonal, vascular, and metabolic abnormalities. These changes trigger a complex biological response aimed at curtailing the physical damage and restoring homeostasis and functionality. Although a positive correlation exists between the type and severity of TBI and PTE, there is only an incomplete understanding of the time-dependent sequelae of TBI pathobiologies and their role in epileptogenesis. Determining the temporal profile of protein biomarkers in the blood (serum or plasma) and cerebrospinal fluid (CSF) can help to identify pathobiologies underlying the development of PTE, high-risk individuals, and disease modifying therapies. Here we review the pathobiological sequelae of TBI in the context of blood- and CSF-based protein biomarkers, their potential role in epileptogenesis, and discuss future directions aimed at improving the diagnosis and treatment of PTE.
Collapse
Affiliation(s)
- Denes V Agoston
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, USA.
| | - Alaa Kamnaksh
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, USA
| |
Collapse
|
29
|
Gasperi V, Sibilano M, Savini I, Catani MV. Niacin in the Central Nervous System: An Update of Biological Aspects and Clinical Applications. Int J Mol Sci 2019; 20:ijms20040974. [PMID: 30813414 PMCID: PMC6412771 DOI: 10.3390/ijms20040974] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022] Open
Abstract
Niacin (also known as "vitamin B₃" or "vitamin PP") includes two vitamers (nicotinic acid and nicotinamide) giving rise to the coenzymatic forms nicotinamide adenine dinucleotide (NAD) and nicotinamide adenine dinucleotide phosphate (NADP). The two coenzymes are required for oxidative reactions crucial for energy production, but they are also substrates for enzymes involved in non-redox signaling pathways, thus regulating biological functions, including gene expression, cell cycle progression, DNA repair and cell death. In the central nervous system, vitamin B₃ has long been recognized as a key mediator of neuronal development and survival. Here, we will overview available literature data on the neuroprotective role of niacin and its derivatives, especially focusing especially on its involvement in neurodegenerative diseases (Alzheimer's, Parkinson's, and Huntington's diseases), as well as in other neuropathological conditions (ischemic and traumatic injuries, headache and psychiatric disorders).
Collapse
Affiliation(s)
- Valeria Gasperi
- Department of Experimental Medicine, Tor Vergata University of Rome, Via Montpellier 1, 00133 Rome, Italy.
| | - Matteo Sibilano
- Department of Experimental Medicine, Tor Vergata University of Rome, Via Montpellier 1, 00133 Rome, Italy.
| | - Isabella Savini
- Department of Experimental Medicine, Tor Vergata University of Rome, Via Montpellier 1, 00133 Rome, Italy.
| | - Maria Valeria Catani
- Department of Experimental Medicine, Tor Vergata University of Rome, Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
30
|
Klyuyeva A, Tuganova A, Kedishvili N, Popov KM. Tissue-specific kinase expression and activity regulate flux through the pyruvate dehydrogenase complex. J Biol Chem 2018; 294:838-851. [PMID: 30482839 DOI: 10.1074/jbc.ra118.006433] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/23/2018] [Indexed: 01/15/2023] Open
Abstract
The pyruvate dehydrogenase complex (PDC) is a multienzyme assembly that converts pyruvate to acetyl-CoA. As pyruvate and acetyl-CoA play central roles in cellular metabolism, understanding PDC regulation is pivotal to understanding the larger metabolic network. The activity of mammalian PDC is regulated through reversible phosphorylation governed by at least four isozymes of pyruvate dehydrogenase kinase (PDK). Deciphering which kinase regulates PDC in organisms at specific times or places has been challenging. In this study, we analyzed mouse strains carrying targeted mutations of individual isozymes to explore their role in regulating PDC activity. Analysis of protein content of PDK isozymes in major metabolic tissues revealed that PDK1 and PDK2 were ubiquitously expressed, whereas PDK3 and PDK4 displayed a rather limited tissue distribution. Measurement of kinase activity showed that PDK1 is the principal isozyme regulating hepatic PDC. PDK2 was largely responsible for inactivation of PDC in tissues of muscle origin and brown adipose tissue (BAT). PDK3 was the principal kinase regulating pyruvate dehydrogenase activity in kidney and brain. In a well-fed state, the tissue levels of PDK4 protein were fairly low. In most tissues tested, PDK4 ablation had little effect on the overall rates of inactivation of PDC in kinase reaction. Taken together, these data strongly suggest that the activity of PDC is regulated by different isozymes in different tissues. Furthermore, it appears that the overall flux through PDC in a given tissue largely reflects the properties of the PDK isozyme that is principally responsible for the regulation of PDC activity in that tissue.
Collapse
Affiliation(s)
- Alla Klyuyeva
- From the Department of Biochemistry and Molecular Genetics, Schools of Medicine and Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Alina Tuganova
- From the Department of Biochemistry and Molecular Genetics, Schools of Medicine and Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Natalia Kedishvili
- From the Department of Biochemistry and Molecular Genetics, Schools of Medicine and Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Kirill M Popov
- From the Department of Biochemistry and Molecular Genetics, Schools of Medicine and Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
31
|
Westermaier T, Stetter C, Koehler D, Weiland J, Lilla N. Acute reaction of arterial blood vessels after experimental subarachnoid hemorrhage - An in vivo microscopic study. J Neurol Sci 2018; 396:172-177. [PMID: 30472554 DOI: 10.1016/j.jns.2018.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/13/2018] [Accepted: 11/05/2018] [Indexed: 11/29/2022]
Abstract
Subarachnoid hemorrhage (SAH) results in a rapid decrease of cerebral perfusion. While cerebral perfusion pressure (CPP) may quickly recover, a sustained decrease of cerebral blood flow (CBF) has been observed. Acute vasospasm has been concluded from this mismatch. This study was conducted to visualize and investigate immediate vascular reactions during and after experimental SAH. Male Sprague-Dawley rats were subjected to SAH by the endovascular filament model (n = 7) or served as controls (n = 4). Videomicroscopy was performed via a cranial window. Regions of interest were defined in areas covered by videomicroscopy and arterial diameters measured at defined time-points from 15 min before until 3 h after SAH. Local CBF was monitored over the opposite hemisphere by laser-Doppler flowmetry. Local CBF showed a typical decrease immediately after vessel perforation followed by an incomplete recovery in the 3 h thereafter. Videomicroscopy demonstrated a sharp decrease of the arterial diameter in the first minutes after SAH. In some animals, SAH was followed by a complete disappearance of arterial vessel filling. In the following minutes, arterial filling reappeared or improved, respectively. All animals subjected to SAH showed significant vasospasm in subarachnoid arteries. This is the first study to visualize acute vascular reactions during and immediately after SAH. Although the cranial window technique only covers a part of the cerebral vasculature, it covers cerebral vessels rather distant from the site of endovascular perforation. Therefore, it is likely that acute vasospasm observed in the monitored areas reflects a global vascular reaction.
Collapse
Affiliation(s)
- Thomas Westermaier
- Department of Neurosurgery, University Hospital Wuerzburg, 97080 Wuerzburg, Germany.
| | - Christian Stetter
- Department of Neurosurgery, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Diana Koehler
- Department of Neurosurgery, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Judith Weiland
- Department of Neurosurgery, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Nadine Lilla
- Department of Neurosurgery, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| |
Collapse
|
32
|
|
33
|
Fan Y, Xiao S. Progression Rate Associated Peripheral Blood Biomarkers of Parkinson's Disease. J Mol Neurosci 2018; 65:312-318. [PMID: 29936662 DOI: 10.1007/s12031-018-1102-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/14/2018] [Indexed: 10/28/2022]
Abstract
Parkinson disease (PD) is one of the most frequent neurodegenerative disorders. The aim of this study was to identify blood biomarkers capable to discriminate PD patients with different progression rates. Differentially expressed genes (DEGs) were acquired by comparing the expression profiles of PD patients with rapid and slow progression rates, using an expression dataset from Gene Expression Omnibus (GEO) under accession code of GSE80599. Altered biological processes and pathways were revealed by functional annotation. Potential biomarkers of PD were identified by protein-protein interaction (PPI) network analysis. Critical transcription factors (TFs) and miRNAs regulating DEGs were predicted by TF analysis and miRNA analysis. A total of 225 DEGs were identified between PD patients with rapid and slow progression profiles. These genes were significantly enriched in biological processes and pathways related to fatty acid metabolism. Among these DEGs, ZFAND4, SRMS, UBL4B, PVALB, DIRAS1, PDP2, LRCH1, and MYL4 were potential progression rate associated biomarkers of PD. Additionally, these DEGs may be regulated by miRNAs of the miR-30 family and TFs STAT1 and GRHL3. Our results may contribute to our understanding of the molecular mechanisms underlying different PD progression profiles.
Collapse
Affiliation(s)
- Yanxia Fan
- Department of Neurology, Fourth People's Hospital of Jinan, No. 50 Shifan Road, Tianqiao District, Jinan, 250031, Shandong Province, China
| | - Shuping Xiao
- Department of Neurology, Fourth People's Hospital of Jinan, No. 50 Shifan Road, Tianqiao District, Jinan, 250031, Shandong Province, China.
| |
Collapse
|
34
|
Dimer NW, Ferreira BK, Agostini JF, Gomes ML, Kist LW, Malgarin F, Carvalho-Silva M, Gomes LM, Rebelo J, Frederico MJS, Silva FRMB, Rico EP, Bogo MR, Streck EL, Ferreira GC, Schuck PF. Brain bioenergetics in rats with acute hyperphenylalaninemia. Neurochem Int 2018; 117:188-203. [PMID: 29454001 DOI: 10.1016/j.neuint.2018.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 12/21/2017] [Accepted: 01/03/2018] [Indexed: 12/15/2022]
Abstract
Phenylketonuria (PKU) is a disorder of phenylalanine (Phe) metabolism caused by deficient phenylalanine hydroxylase (PAH) activity. The deficiency results in increased levels of Phe and its metabolites in fluids and tissues of patients. PKU patients present neurological signs and symptoms including hypomyelination and intellectual deficit. This study assessed brain bioenergetics at 1 h after acute Phe administration to induce hyperphenylalaninemia (HPA) in rats. Wistar rats were randomized in two groups: HPA animals received a single subcutaneous administration of Phe (5.2 μmol/g) plus p-Cl-Phe (PAH inhibitor) (0.9 μmol/g); control animals received a single injection of 0.9% NaCl. In cerebral cortex, HPA group showed lower mitochondrial mass, lower glycogen levels, as well as lower activities of complexes I-III and IV, ATP synthase and citrate synthase. Higher levels of free Pi and phospho-AMPK, and higher activities of LDH, α-ketoglutarate dehydrogenase and isocitrate dehydrogenase were also reported in cerebral cortex of HPA animals. In striatum, HPA animals had higher LDH (pyruvate to lactate) and isocitrate dehydrogenase activities, and lower activities of α-ketoglutarate dehydrogenase and complex IV, as well as lower phospho-AMPK immunocontent. In hippocampus, HPA rats had higher mRNA expression for MFN1 and higher activities of α-ketoglutarate dehydrogenase and isocitrate dehydrogenase, but decreased activities of pyruvate dehydrogenase and complexes I and IV. In conclusion, our data demonstrated impaired bioenergetics in cerebral cortex, striatum and hippocampus of HPA rats.
Collapse
Affiliation(s)
- Nádia Weber Dimer
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Bruna Klippel Ferreira
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil; Laboratório de Neuroenergética e Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jotele Fontana Agostini
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Maria Luiza Gomes
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Luiza Wilges Kist
- Laboratório de Biologia Genômica e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Malgarin
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Milena Carvalho-Silva
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Lara Mezari Gomes
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Joyce Rebelo
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Marisa Jádna Silva Frederico
- Laboratório de Hormônios e Transdução de Sinais, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Fátima Regina Mena Barreto Silva
- Laboratório de Hormônios e Transdução de Sinais, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Eduardo Pacheco Rico
- Laboratório de Sinalização Neural e Psicofarmacologia, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Mauricio Reis Bogo
- Laboratório de Biologia Genômica e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Emilio Luiz Streck
- Laboratório de Bioenergética, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gustavo Costa Ferreira
- Laboratório de Neuroenergética e Erros Inatos do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patrícia Fernanda Schuck
- Laboratório de Erros Inatos do Metabolismo, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.
| |
Collapse
|
35
|
Synergistic Association of Valproate and Resveratrol Reduces Brain Injury in Ischemic Stroke. Int J Mol Sci 2018; 19:ijms19010172. [PMID: 29316653 PMCID: PMC5796121 DOI: 10.3390/ijms19010172] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/15/2017] [Accepted: 01/02/2018] [Indexed: 11/16/2022] Open
Abstract
Histone deacetylation, together with altered acetylation of NF-κB/RelA, encompassing the K310 residue acetylation, occur during brain ischemia. By restoring the normal acetylation condition, we previously reported that sub-threshold doses of resveratrol and entinostat (MS-275), respectively, an activator of the AMP-activated kinase (AMPK)-sirtuin 1 pathway and an inhibitor of class I histone deacetylases (HDACs), synergistically elicited neuroprotection in a mouse model of ischemic stroke. To improve the translational power of this approach, we investigated the efficacy of MS-275 replacement with valproate, the antiepileptic drug also reported to be a class I HDAC blocker. In cortical neurons previously exposed to oxygen glucose deprivation (OGD), valproate elicited neuroprotection at 100 nmol/mL concentration when used alone and at 1 nmol/mL concentration when associated with resveratrol (3 nmol/mL). Resveratrol and valproate restored the acetylation of histone H3 (K9/18), and they reduced the RelA(K310) acetylation and the Bim level in neurons exposed to OGD. Chromatin immunoprecipitation analysis showed that the synergistic drug association impaired the RelA binding to the Bim promoter, as well as the promoter-specific H3 (K9/18) acetylation. In mice subjected to 60 min of middle cerebral artery occlusion (MCAO), the association of resveratrol 680 µg/kg and valproate 200 µg/kg significantly reduced the infarct volume as well as the neurological deficits. The present study suggests that valproate and resveratrol may represent a promising ready-to-use strategy to treat post-ischemic brain damage.
Collapse
|
36
|
Zhao L, Zhang Z, Zhou M, Gou X, Zeng Y, Song J, Ma W, Xu Y. A urinary metabolomics (GC-MS) strategy to evaluate the antidepressant-like effect of chlorogenic acid in adrenocorticotropic hormone-treated rats. RSC Adv 2018; 8:9141-9151. [PMID: 35541857 PMCID: PMC9078588 DOI: 10.1039/c8ra00074c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/14/2018] [Indexed: 11/25/2022] Open
Abstract
Major depressive disorder (MDD) is a chronic recurring illness that seriously affects human health. Chlorogenic acid (CGA), an important polyphenol extracted from Eucommia ulmoides Oliver bark, has been reported to have anti-depression, neuroprotection, memory improvement and other pharmacological effects. However, little is known about the underlying mechanisms of CGA on the treatment of depression. Here, we investigated the antidepressant-like effects of CGA on an adrenocorticotropic hormone (ACTH)-treated rat model. Thirty-two male Wistar rats were randomly divided into four groups: normal diet group (N), ACTH-treated model group (M), memantine positive control group (M + Mem) and CGA intervened group (M + CGA). Sucrose preference tests (SPTs) and open-field tests (OFTs) were performed to evaluate depressive-like behaviors. Memantine (30 mg kg−1) and CGA (500 mg kg−1) administration dramatically increased hedonic behaviors of the rats in SPT. The scores of crossing and rearing were significantly increased in the M + Mem group and M + CGA group. These results of the behaviour tests might be suggestive of antidepressant-like effects. Moreover, memantine and CGA reversed the levels of serum 5-hydroxytryptamine (5-HT), ACTH, corticotropin-releasing hormone (CRH), and dopamine (DA) that were altered in ACTH-treated rats. Based on a GC-MS metabolomic approach, significant differences in the metabolic profile were observed in ACTH-treated rats compared with the control group, as well as the M + CGA group and M + Mem group compared with the ACTH-treated group. A total of 19 metabolites were identified for the discrimination of normal rats and ACTH-treated rats, and 12 out of 19 differential metabolites were reversed with CGA intervention. Combined with pattern recognition and bioinformatics, nine perturbed metabolic pathways, including energy metabolism, neurotransmitter metabolism, and amino acid metabolism, were identified based on these metabolites. These integrative studies might give a holistic insight into the pathophysiological mechanism of the ACTH-treated depressive rat model, and also showed that CGA has antidepressant-like activities in ACTH-treated rats, providing an important drug candidate for the prevention and treatment of tricyclic anti-depressant treatment-resistant depression. Chlorogenic acid showed antidepressant-like activity in chronic ACTH-treated rats, providing a potential drug candidate for prevention and treatment of tricyclic antidepressant treatment-resistant depression. Related metabolic pathways were shown.![]()
Collapse
Affiliation(s)
- Le Zhao
- Center for Chinese Medicine Therapy and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| | - Zixu Zhang
- Center for Chinese Medicine Therapy and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
- College of Chinese Pharmacy
| | - Mingmei Zhou
- Center for Chinese Medicine Therapy and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
- Key Laboratory of Medicinal Animal and Plant Resources in Qinghai-Tibet Plateau
| | - Xiaojun Gou
- Central Laboratory
- Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201999
- China
| | - Yang Zeng
- College of Life Science
- Qinghai Normal University
- Xining
- China
- Key Laboratory of Medicinal Animal and Plant Resources in Qinghai-Tibet Plateau
| | - Jing Song
- Center for Chinese Medicine Therapy and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| | - Weini Ma
- Center for Chinese Medicine Therapy and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| | - Ying Xu
- Department of Physiology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| |
Collapse
|
37
|
Liu L, MacKenzie KR, Putluri N, Maletić-Savatić M, Bellen HJ. The Glia-Neuron Lactate Shuttle and Elevated ROS Promote Lipid Synthesis in Neurons and Lipid Droplet Accumulation in Glia via APOE/D. Cell Metab 2017; 26:719-737.e6. [PMID: 28965825 PMCID: PMC5677551 DOI: 10.1016/j.cmet.2017.08.024] [Citation(s) in RCA: 316] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/21/2017] [Accepted: 08/30/2017] [Indexed: 01/04/2023]
Abstract
Elevated reactive oxygen species (ROS) induce the formation of lipids in neurons that are transferred to glia, where they form lipid droplets (LDs). We show that glial and neuronal monocarboxylate transporters (MCTs), fatty acid transport proteins (FATPs), and apolipoproteins are critical for glial LD formation. MCTs enable glia to secrete and neurons to absorb lactate, which is converted to pyruvate and acetyl-CoA in neurons. Lactate metabolites provide a substrate for synthesis of fatty acids, which are processed and transferred to glia by FATP and apolipoproteins. In the presence of high ROS, inhibiting lactate transfer or lowering FATP or apolipoprotein levels decreases glial LD accumulation in flies and in primary mouse glial-neuronal cultures. We show that human APOE can substitute for a fly glial apolipoprotein and that APOE4, an Alzheimer's disease susceptibility allele, is impaired in lipid transport and promotes neurodegeneration, providing insights into disease mechanisms.
Collapse
Affiliation(s)
- Lucy Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin R MacKenzie
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology and Advanced Technology Cor, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletić-Savatić
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
38
|
Piao L, Fang YH, Kubler MM, Donnino MW, Sharp WW. Enhanced pyruvate dehydrogenase activity improves cardiac outcomes in a murine model of cardiac arrest. PLoS One 2017; 12:e0185046. [PMID: 28934276 PMCID: PMC5608301 DOI: 10.1371/journal.pone.0185046] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/04/2017] [Indexed: 11/19/2022] Open
Abstract
Rationale Post-ischemic changes in cellular metabolism alter myocardial and neurological function. Pyruvate dehydrogenase (PDH), the limiting step in mitochondrial glucose oxidation, is inhibited by increased expression of PDH kinase (PDK) during ischemia/reperfusion injury. This results in decreased utilization of glucose to generate cellular ATP. Post-cardiac arrest (CA) hypothermia improves outcomes and alters metabolism, but its influence on PDH and PDK activity following CA are unknown. We hypothesized that therapeutic hypothermia (TH) following CA is associated with the inhibition of PDK activity and increased PDH activity. We further hypothesized that an inhibitor of PDK activity, dichloroacetate (DCA), would improve PDH activity and post-CA outcomes. Methods and results Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent a 12-minute KCl-induced CA followed by cardiopulmonary resuscitation. Compared to normothermic (37°C) CA controls, administering TH (30°C) improved overall survival (72-hour survival rate: 62.5% vs. 28.6%, P<0.001), post-resuscitation myocardial function (ejection fraction: 50.9±3.1% vs. 27.2±2.0%, P<0.001; aorta systolic pressure: 132.7±7.3 vs. 72.3±3.0 mmHg, P<0.001), and neurological scores at 72-hour post CA (9.5±1.3 vs. 5.4±1.3, P<0.05). In both heart and brain, CA increased lactate concentrations (1.9-fold and 3.1-fold increase, respectively, P<0.01), decreased PDH enzyme activity (24% and 50% reduction, respectively, P<0.01), and increased PDK protein expressions (1.2-fold and 1.9-fold, respectively, P<0.01). In contrast, post-CA treatment with TH normalized lactate concentrations (P<0.01 and P<0.05) and PDK expressions (P<0.001 and P<0.05), while increasing PDH activity (P<0.01 and P<0.01) in both the heart and brain. Additionally, treatment with DCA (0.2 mg/g body weight) 30 min prior to CA improved both myocardial hemodynamics 2 hours post-CA (aortic systolic pressure: 123±3 vs. 96±4 mmHg, P<0.001) and 72-hour survival rates (50% vs. 19%, P<0.05) in normothermic animals. Conclusions Enhanced PDH activity in the setting of TH or DCA administration is associated with improved post-CA resuscitation outcomes. PDH is a promising therapeutic target for improving post-CA outcomes.
Collapse
Affiliation(s)
- Lin Piao
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Yong-Hu Fang
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Manfred M. Kubler
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Michael W. Donnino
- Departments of Emergency Medicine and Internal Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Willard W. Sharp
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
39
|
Madeira JP, Alpha-Bazin BM, Armengaud J, Duport C. Methionine Residues in Exoproteins and Their Recycling by Methionine Sulfoxide Reductase AB Serve as an Antioxidant Strategy in Bacillus cereus. Front Microbiol 2017; 8:1342. [PMID: 28798727 PMCID: PMC5526929 DOI: 10.3389/fmicb.2017.01342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/03/2017] [Indexed: 11/13/2022] Open
Abstract
During aerobic respiratory growth, Bacillus cereus is exposed to continuously reactive oxidant, produced by partially reduced forms of molecular oxygen, known as reactive oxygen species (ROS). The sulfur-containing amino acid, methionine (Met), is particularly susceptible to ROS. The major oxidation products, methionine sulfoxides, can be readily repaired by methionine sulfoxide reductases, which reduce methionine sulfoxides [Met(O)] back to methionine. Here, we show that methionine sulfoxide reductase AB (MsrAB) regulates the Met(O) content of both the cellular proteome and exoproteome of B. cereus in a growth phase-dependent manner. Disruption of msrAB leads to metabolism changes resulting in enhanced export of Met(O) proteins at the late exponential growth phase and enhanced degradation of exoproteins. This suggests that B. cereus can modulate its capacity and specificity for protein export/secretion through the growth phase-dependent expression of msrAB. Our results also show that cytoplasmic MsrAB recycles Met residues in enterotoxins, which are major virulence factors in B. cereus.
Collapse
Affiliation(s)
- Jean-Paul Madeira
- Sécurité et Qualité des Produits d'Origine Végétale (SQPOV), UMR0408, Avignon Université, Institut National de la Recherche AgronomiqueAvignon, France.,Commissariat à lEnergie Atomique, Direction de la Recherche Fondamentale, Institut des Sciences du vivant Frédéric-Joliot (Joliot), Service de Pharmacologie et Immunoanalyse, Laboratoire Innovations Technologiques pour la Détection et le Diagnostic (Li2D)Bagnols-sur-Cèze, France
| | - Béatrice M Alpha-Bazin
- Commissariat à lEnergie Atomique, Direction de la Recherche Fondamentale, Institut des Sciences du vivant Frédéric-Joliot (Joliot), Service de Pharmacologie et Immunoanalyse, Laboratoire Innovations Technologiques pour la Détection et le Diagnostic (Li2D)Bagnols-sur-Cèze, France
| | - Jean Armengaud
- Commissariat à lEnergie Atomique, Direction de la Recherche Fondamentale, Institut des Sciences du vivant Frédéric-Joliot (Joliot), Service de Pharmacologie et Immunoanalyse, Laboratoire Innovations Technologiques pour la Détection et le Diagnostic (Li2D)Bagnols-sur-Cèze, France
| | - Catherine Duport
- Sécurité et Qualité des Produits d'Origine Végétale (SQPOV), UMR0408, Avignon Université, Institut National de la Recherche AgronomiqueAvignon, France
| |
Collapse
|
40
|
L-Carnitine and Acetyl-L-carnitine Roles and Neuroprotection in Developing Brain. Neurochem Res 2017; 42:1661-1675. [PMID: 28508995 DOI: 10.1007/s11064-017-2288-7] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/30/2022]
Abstract
L-Carnitine functions to transport long chain fatty acyl-CoAs into the mitochondria for degradation by β-oxidation. Treatment with L-carnitine can ameliorate metabolic imbalances in many inborn errors of metabolism. In recent years there has been considerable interest in the therapeutic potential of L-carnitine and its acetylated derivative acetyl-L-carnitine (ALCAR) for neuroprotection in a number of disorders including hypoxia-ischemia, traumatic brain injury, Alzheimer's disease and in conditions leading to central or peripheral nervous system injury. There is compelling evidence from preclinical studies that L-carnitine and ALCAR can improve energy status, decrease oxidative stress and prevent subsequent cell death in models of adult, neonatal and pediatric brain injury. ALCAR can provide an acetyl moiety that can be oxidized for energy, used as a precursor for acetylcholine, or incorporated into glutamate, glutamine and GABA, or into lipids for myelination and cell growth. Administration of ALCAR after brain injury in rat pups improved long-term functional outcomes, including memory. Additional studies are needed to better explore the potential of L-carnitine and ALCAR for protection of developing brain as there is an urgent need for therapies that can improve outcome after neonatal and pediatric brain injury.
Collapse
|
41
|
Pavón N, Cabrera-Orefice A, Gallardo-Pérez JC, Uribe-Alvarez C, Rivero-Segura NA, Vazquez-Martínez ER, Cerbón M, Martínez-Abundis E, Torres-Narvaez JC, Martínez-Memije R, Roldán-Gómez FJ, Uribe-Carvajal S. In female rat heart mitochondria, oophorectomy results in loss of oxidative phosphorylation. J Endocrinol 2017; 232:221-235. [PMID: 27872198 DOI: 10.1530/joe-16-0161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 11/21/2016] [Indexed: 01/13/2023]
Abstract
Oophorectomy in adult rats affected cardiac mitochondrial function. Progression of mitochondrial alterations was assessed at one, two and three months after surgery: at one month, very slight changes were observed, which increased at two and three months. Gradual effects included decrease in the rates of oxygen consumption and in respiratory uncoupling in the presence of complex I substrates, as well as compromised Ca2+ buffering ability. Malondialdehyde concentration increased, whereas the ROS-detoxifying enzyme Mn2+ superoxide dismutase (MnSOD) and aconitase lost activity. In the mitochondrial respiratory chain, the concentration and activity of complex I and complex IV decreased. Among other mitochondrial enzymes and transporters, adenine nucleotide carrier and glutaminase decreased. 2-Oxoglutarate dehydrogenase and pyruvate dehydrogenase also decreased. Data strongly suggest that in the female rat heart, estrogen depletion leads to progressive, severe mitochondrial dysfunction.
Collapse
Affiliation(s)
- Natalia Pavón
- Departamento de FarmacologíaInstituto Nacional de Cardiología Ignacio Chávez, México, Mexico
| | - Alfredo Cabrera-Orefice
- Departamento de Genética MolecularInstituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| | | | - Cristina Uribe-Alvarez
- Departamento de Genética MolecularInstituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| | - Nadia A Rivero-Segura
- Unidad de Investigación en Reproducción HumanaInstituto Nacional de Perinatología-Facultad de Química UNAM, México D.F., Mexico
| | - Edgar Ricardo Vazquez-Martínez
- Unidad de Investigación en Reproducción HumanaInstituto Nacional de Perinatología-Facultad de Química UNAM, México D.F., Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción HumanaInstituto Nacional de Perinatología-Facultad de Química UNAM, México D.F., Mexico
| | - Eduardo Martínez-Abundis
- División Académica Multidisciplinaria de ComalcalcoUniversidad Juárez Autónoma de Tabasco, México, Mexico
| | | | - Raúl Martínez-Memije
- Departamento de Instrumentación ElectromecánicaInstituto Nacional de Cardiología Ignacio Chávez, Tlalpan DF, México, Mexico
| | | | - Salvador Uribe-Carvajal
- Departamento de Genética MolecularInstituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| |
Collapse
|
42
|
Lai YW, Wu SB, Hsueh TY, Chiu AW, Wei YH, Chen SSS. Enhanced oxidative stress and the glycolytic switch in superficial urothelial carcinoma of urinary bladder. UROLOGICAL SCIENCE 2016. [DOI: 10.1016/j.urols.2015.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
43
|
Dezfulian C, Kenny E, Lamade A, Misse A, Krehel N, St Croix C, Kelley EE, Jackson TC, Uray T, Rackley J, Kochanek PM, Clark RSB, Bayir H. Mechanistic characterization of nitrite-mediated neuroprotection after experimental cardiac arrest. J Neurochem 2016; 139:419-431. [PMID: 27507435 DOI: 10.1111/jnc.13764] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 12/27/2022]
Abstract
Nitrite acts as an ischemic reservoir of nitric oxide (NO) and a potent S-nitrosating agent which reduced histologic brain injury after rat asphyxial cardiac arrest (ACA). The mechanism(s) of nitrite-mediated neuroprotection remain to be defined. We hypothesized that nitrite-mediated brain mitochondrial S-nitrosation accounts for neuroprotection by reducing reperfusion reactive oxygen species (ROS) generation. Nitrite (4 μmol) or placebo was infused IV after normothermic (37°C) ACA in randomized, blinded fashion with evaluation of neurologic function, survival, brain mitochondrial function, and ROS. Blood and CSF nitrite were quantified using reductive chemiluminescence and S-nitrosation by biotin switch. Direct neuroprotection was verified in vitro after 1 and 4 h neuronal oxygen glucose deprivation measuring neuronal death with inhibition studies to examine mechanism. Mitochondrial ROS generation was quantified by live neuronal imaging using mitoSOX. Nitrite significantly reduced neurologic disability after ACA. ROS generation was reduced in brain mitochondria from nitrite- versus placebo-treated rats after ACA with congruent preservation of brain ascorbate and reduction of ROS in brain sections using immuno-spin trapping. ATP generation was maintained with nitrite up to 24 h after ACA. Nitrite rapidly entered CSF and increased brain mitochondrial S-nitrosation. Nitrite reduced in vitro mitochondrial superoxide generation and improved survival of neurons after oxygen glucose deprivation. Protection was maintained with inhibition of soluble guanylate cyclase but lost with NO scavenging and ultraviolet irradiation. Nitrite therapy results in direct neuroprotection from ACA mediated by reductions in brain mitochondrial ROS in association with protein S-nitrosation. Neuroprotection is dependent on NO and S-nitrosothiol generation, not soluble guanylate cyclase.
Collapse
Affiliation(s)
- Cameron Dezfulian
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Elizabeth Kenny
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrew Lamade
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amalea Misse
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicholas Krehel
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Claudette St Croix
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Eric E Kelley
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Travis C Jackson
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas Uray
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Justin Rackley
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert S B Clark
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hulya Bayir
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
44
|
1'-Acetoxychavicol acetate ameliorates age-related spatial memory deterioration by increasing serum ketone body production as a complementary energy source for neuronal cells. Chem Biol Interact 2016; 257:101-9. [PMID: 27481192 DOI: 10.1016/j.cbi.2016.07.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/25/2016] [Accepted: 07/27/2016] [Indexed: 11/24/2022]
Abstract
1'-Acetoxychavicol acetate (ACA) is naturally obtained from the rhizomes and seeds of Alpinia galangal. Here, we examined the effect of ACA on learning and memory in senescence-accelerated mice prone 8 (SAMP8). In mice that were fed a control diet containing 0.02% ACA for 25 weeks, the learning ability in the Morris water maze test was significantly enhanced in comparison with mice that were fed the control diet alone. In the Y-maze test, SAMP8 mice showed decreased spontaneous alterations in comparison with senescence-accelerated resistant/1 (SAMR1) mice, a homologous control, which was improved by ACA pretreatment. Serum metabolite profiles were obtained by GC-MS analysis, and each metabolic profile was plotted on a 3D score plot. Based upon the diagram, it can be seen that the distribution areas for the three groups were completely separate. Furthermore, the contents of β-hydroxybutyric acid and palmitic acid in the serum of SAMP8-ACA mice were higher than those of SAMP8-control mice and SAMR1-control mice. We also found that SAMR1 mice did not show histological abnormalities, whereas histological damage in the CA1 region of the hippocampus in SAMP8-control mice was observed. However, SAMP8-ACA mice were observed in a similar manner as SAMR1 mice. These findings confirm that ACA increases the serum concentrations of β-hydroxybutyric acid and palmitic acid levels and thus these fuels might contribute to the maintenance of the cognitive performance of SAMP8 mice.
Collapse
|
45
|
Thibodeau A, Geng X, Previch LE, Ding Y. Pyruvate dehydrogenase complex in cerebral ischemia-reperfusion injury. Brain Circ 2016; 2:61-66. [PMID: 30276274 PMCID: PMC6126256 DOI: 10.4103/2394-8108.186256] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 06/02/2016] [Accepted: 06/14/2016] [Indexed: 11/11/2022] Open
Abstract
Pyruvate dehydrogenase (PDH) complex is a mitochondrial matrix enzyme that serves a critical role in the conversion of anaerobic to aerobic cerebral energy. The regulatory complexity of PDH, coupled with its significant influence in brain metabolism, underscores its susceptibility to, and significance in, ischemia-reperfusion injury. Here, we evaluate proposed mechanisms of PDH-mediated neurodysfunction in stroke, including oxidative stress, altered regulatory enzymatic control, and loss of PDH activity. We also describe the neuroprotective influence of antioxidants, dichloroacetate, acetyl-L-carnitine, and combined therapy with ethanol and normobaric oxygen, explained in relation to PDH modulation. Our review highlights the significance of PDH impairment in stroke injury through an understanding of the mechanisms by which it is modulated, as well as an exploration of neuroprotective strategies available to limit its impairment.
Collapse
Affiliation(s)
- Alexa Thibodeau
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA.,China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Lauren E Previch
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuchuan Ding
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA.,China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
46
|
Shahjouei S, Ansari S, Pourmotabbed T, Zand R. Potential Roles of Adropin in Central Nervous System: Review of Current Literature. Front Mol Biosci 2016; 3:25. [PMID: 27446928 PMCID: PMC4921473 DOI: 10.3389/fmolb.2016.00025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/27/2016] [Indexed: 01/04/2023] Open
Abstract
Adropin is a 4.9 kDa peptide that is important for maintenance of metabolic and non-metabolic homeostasis. It regulates glucose and fatty acid metabolism and is involved in endothelial cell function and endothelial nitric oxide (NO) synthase bioactivity as well as physical activity and motor coordination. Adropin is expressed in many tissues and organs including central nervous system (CNS). This peptide plays a crucial role in the development of various CNS disorders such as stroke, schizophrenia, bipolar disorder as well as Alzheimer's, Parkinson's, and Huntington's diseases. In this comprehensive review, the potential roles of adropin in cellular signaling pathways that lead to pathogenesis and/or treatment of CNS disorders will be discussed.
Collapse
Affiliation(s)
- Shima Shahjouei
- Department of Neurosurgery, Tehran University of Medical Sciences Tehran, Iran
| | - Saeed Ansari
- Department of Neurology, University of Tennessee Health Science Center Memphis, TN, USA
| | - Tayebeh Pourmotabbed
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center Memphis, TN, USA
| | - Ramin Zand
- Department of Neurology, University of Tennessee Health Science CenterMemphis, TN, USA; Biocomplexity Institute, Virginia Polytechnic Institute and State UniversityBlacksburg, VA, USA
| |
Collapse
|
47
|
Ikeda K, Liu X, Kida K, Marutani E, Hirai S, Sakaguchi M, Andersen LW, Bagchi A, Cocchi MN, Berg KM, Ichinose F, Donnino MW. Thiamine as a neuroprotective agent after cardiac arrest. Resuscitation 2016; 105:138-44. [PMID: 27185216 DOI: 10.1016/j.resuscitation.2016.04.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/19/2016] [Accepted: 04/25/2016] [Indexed: 11/19/2022]
Abstract
AIMS Reduction of pyruvate dehydrogenase (PDH) activity in the brain is associated with neurological deficits in animals resuscitated from cardiac arrest. Thiamine is an essential co-factor of PDH. The objective of this study was to examine whether administration of thiamine improves outcomes after cardiac arrest in mice. Secondarily, we aimed to characterize the impact of cardiac arrest on PDH activity in mice and humans. METHODS Animal study: Adult mice were subjected to cardiac arrest whereupon cardiopulmonary resuscitation was performed. Thiamine or vehicle was administered 2min before resuscitation and daily thereafter. Mortality, neurological outcome, and metabolic markers were evaluated. Human study: In a convenience sample of post-cardiac arrest patients, we measured serial PDH activity from peripheral blood mononuclear cells and compared them to healthy controls. RESULTS Animal study: Mice treated with thiamine had increased 10-day survival (48% versus 17%, P<0.01) and improved neurological function when compared to vehicle-treated mice. In addition, thiamine markedly improved histological brain injury compared to vehicle. The beneficial effects of thiamine were accompanied by improved oxygen consumption in mitochondria, restored thiamine pyrophosphate levels, and increased PDH activity in the brain at 10 days. Human study: Post-cardiac arrest patients had lower PDH activity in mononuclear cells than did healthy volunteers (estimated difference: -5.8O.D./min/mg protein, P<0.001). CONCLUSIONS The provision of thiamine after cardiac arrest improved neurological outcome and 10-day survival in mice. PDH activity was markedly depressed in post-cardiac arrest patients suggesting that this pathway may represent a therapeutic target.
Collapse
Affiliation(s)
- Kohei Ikeda
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Xiaowen Liu
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kotaro Kida
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Eizo Marutani
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Shuichi Hirai
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Masahiro Sakaguchi
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lars W Andersen
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Anaesthesiology, Aarhus University Hospital, Aarhus, Denmark; Research Center for Emergency Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Aranya Bagchi
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Michael N Cocchi
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Anesthesia Critical Care, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Katherine M Berg
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Division of Pulmonary and Critical Care, Beth Israel Deaconess Medical Center, MA, USA
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Michael W Donnino
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Division of Pulmonary and Critical Care, Beth Israel Deaconess Medical Center, MA, USA.
| |
Collapse
|
48
|
Akbar M, Essa MM, Daradkeh G, Abdelmegeed MA, Choi Y, Mahmood L, Song BJ. Mitochondrial dysfunction and cell death in neurodegenerative diseases through nitroxidative stress. Brain Res 2016; 1637:34-55. [PMID: 26883165 PMCID: PMC4821765 DOI: 10.1016/j.brainres.2016.02.016] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 02/02/2016] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Abstract
Mitochondria are important for providing cellular energy ATP through the oxidative phosphorylation pathway. They are also critical in regulating many cellular functions including the fatty acid oxidation, the metabolism of glutamate and urea, the anti-oxidant defense, and the apoptosis pathway. Mitochondria are an important source of reactive oxygen species leaked from the electron transport chain while they are susceptible to oxidative damage, leading to mitochondrial dysfunction and tissue injury. In fact, impaired mitochondrial function is commonly observed in many types of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, alcoholic dementia, brain ischemia-reperfusion related injury, and others, although many of these neurological disorders have unique etiological factors. Mitochondrial dysfunction under many pathological conditions is likely to be promoted by increased nitroxidative stress, which can stimulate post-translational modifications (PTMs) of mitochondrial proteins and/or oxidative damage to mitochondrial DNA and lipids. Furthermore, recent studies have demonstrated that various antioxidants, including naturally occurring flavonoids and polyphenols as well as synthetic compounds, can block the formation of reactive oxygen and/or nitrogen species, and thus ultimately prevent the PTMs of many proteins with improved disease conditions. Therefore, the present review is aimed to describe the recent research developments in the molecular mechanisms for mitochondrial dysfunction and tissue injury in neurodegenerative diseases and discuss translational research opportunities.
Collapse
Affiliation(s)
- Mohammed Akbar
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, College of Agriculture and Marine Sciences, Sultan Qaboos University, Oman; Ageing and Dementia Research Group, Sultan Qaboos University, Oman
| | - Ghazi Daradkeh
- Department of Food Science and Nutrition, College of Agriculture and Marine Sciences, Sultan Qaboos University, Oman
| | - Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Youngshim Choi
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Lubna Mahmood
- Department of Nutritional Sciences, Qatar University, Qatar
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
49
|
Li H, Liu Y, Lin LT, Wang XR, Du SQ, Yan CQ, He T, Yang JW, Liu CZ. Acupuncture reversed hippocampal mitochondrial dysfunction in vascular dementia rats. Neurochem Int 2015; 92:35-42. [PMID: 26682902 DOI: 10.1016/j.neuint.2015.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 11/16/2015] [Accepted: 12/02/2015] [Indexed: 12/19/2022]
Abstract
Hippocampal mitochondrial dysfunction due to oxidative stress has been considered to play a major role in the pathogenesis of vascular dementia (VD). Previous studies suggested that acupuncture could improve cerebral hypoperfusion-induced cognitive impairments. However, whether hippocampal mitochondria are associated with this cognitive improvement remains unclear. In this study, an animal model of VD was established via bilateral common carotid arteries occlusion (BCCAO) to investigate the alterations of cognitive ability and hippocampal mitochondrial function. BCCAO rats showed impairments in hippocampal mitochondrial function, overproduction of reactive oxygen species (ROS) and learning and memory deficits. After two-week acupuncture treatment, BCCAO-induced spatial learning and memory impairments as shown in Morris water maze were ameliorated. Hippocampal mitochondrial respiratory complex enzymes (complex I, II, IV) activities and cytochrome c oxidase IV expression significantly increased, which might contribute to the reduction of hippocampal ROS generation. In addition, acupuncture significantly improve mitochondrial bioenergy parameters such as mitochondrial respiratory control rate and membrane potential not PDH A1 expression. Placebo-acupuncture did not produce similar therapeutic effects. These findings suggested that acupuncture reversed BCCAO-induced hippocampal mitochondrial dysfunction, which might contribute to its prevention on cognitive deficits.
Collapse
Affiliation(s)
- Hui Li
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China; Tianjin University of Traditional Chinese Medicine, 312 Anshanxidao Street, Nankai District, Tianjin 300193, China
| | - Yi Liu
- Tianjin University of Traditional Chinese Medicine, 312 Anshanxidao Street, Nankai District, Tianjin 300193, China
| | - Li-Ting Lin
- Tianjin University of Traditional Chinese Medicine, 312 Anshanxidao Street, Nankai District, Tianjin 300193, China
| | - Xue-Rui Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Si-Qi Du
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Chao-Qun Yan
- Tianjin University of Traditional Chinese Medicine, 312 Anshanxidao Street, Nankai District, Tianjin 300193, China
| | - Tian He
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Jing-Wen Yang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Cun-Zhi Liu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China.
| |
Collapse
|
50
|
Salminen A, Jouhten P, Sarajärvi T, Haapasalo A, Hiltunen M. Hypoxia and GABA shunt activation in the pathogenesis of Alzheimer's disease. Neurochem Int 2015; 92:13-24. [PMID: 26617286 DOI: 10.1016/j.neuint.2015.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022]
Abstract
We have previously observed that the conversion of mild cognitive impairment to definitive Alzheimer's disease (AD) is associated with a significant increase in the serum level of 2,4-dihydroxybutyrate (2,4-DHBA). The metabolic generation of 2,4-DHBA is linked to the activation of the γ-aminobutyric acid (GABA) shunt, an alternative energy production pathway activated during cellular stress, when the function of Krebs cycle is compromised. The GABA shunt can be triggered by local hypoperfusion and subsequent hypoxia in AD brains caused by cerebral amyloid angiopathy. Succinic semialdehyde dehydrogenase (SSADH) is a key enzyme in the GABA shunt, converting succinic semialdehyde (SSA) into succinate, a Krebs cycle intermediate. A deficiency of SSADH activity stimulates the conversion of SSA into γ-hydroxybutyrate (GHB), an alternative route from the GABA shunt. GHB can exert not only acute neuroprotective activities but unfortunately also chronic detrimental effects which may lead to cognitive impairment. Subsequently, GHB can be metabolized to 2,4-DHBA and secreted from the brain. Thus, the activation of the GABA shunt and the generation of GHB and 2,4-DHBA can have an important role in the early phase of AD pathogenesis.
Collapse
Affiliation(s)
- Antero Salminen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland.
| | - Paula Jouhten
- VTT Technical Research Centre of Finland, FIN-00014 Helsinki, Finland; EMBL European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Timo Sarajärvi
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | - Annakaisa Haapasalo
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, Neulaniementie 2, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, P.O. Box 100, FI-70029 KYS, Finland
| | - Mikko Hiltunen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, P.O. Box 100, FI-70029 KYS, Finland; Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| |
Collapse
|