201
|
Protective Effect of Peroxisome Proliferator-Activated Receptor α Activation against Cardiac Ischemia-Reperfusion Injury Is Related to Upregulation of Uncoupling Protein-3. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3539649. [PMID: 26770648 PMCID: PMC4685116 DOI: 10.1155/2016/3539649] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 08/17/2015] [Accepted: 08/25/2015] [Indexed: 11/17/2022]
Abstract
Activation of peroxisome proliferator-activated receptor α (PPARα) confers cardioprotection, while its mechanism remains elusive. We investigated the protective effect of PPARα activation against cardiac ischemia-reperfusion injury in terms of the expression of uncoupling protein (UCP). Myocardial infarct size and UCP expression were measured in rats treated with WY-14643 20 mg/kg, a PPARα ligand, or vehicle. WY-14643 increased UCP3 expression in vivo. Myocardial infarct size was decreased in the WY-14643 group (76 ± 8% versus 42 ± 12%, P<0.05). During reperfusion, the incidence of arrhythmia was higher in the control group compared with the WY-14643 group (9/10 versus 3/10, P<0.05). H9c2 cells were incubated for 24 h with WY-14643 or vehicle. WY-14643 increased UCP3 expression in H9c2 cells. WY-14643 decreased hypoxia-stimulated ROS production. Cells treated with WY-14643 were more resistant to hypoxia-reoxygenation than the untreated cells. Knocking-down UCP3 by siRNA prevented WY-14643 from attenuating the production of ROS. UCP3 siRNA abolished the effect of WY-14643 on cell viability against hypoxia-reoxygenation. In summary, administration of PPARα agonist WY-14643 mitigated the extent of myocardial infarction and incidence of reperfusion-induced arrhythmia. PPARα activation conferred cytoprotective effect against hypoxia-reoxygenation. Associated mechanisms involved increased UCP3 expression and resultant attenuation of ROS production.
Collapse
|
202
|
Vimaleswaran KS, Cavadino A, Verweij N, Nolte IM, Mateo Leach I, Auvinen J, Veijola J, Elliott P, Penninx BW, Snieder H, Järvelin MR, van der Harst P, Cohen RD, Boucher BJ, Hyppönen E. Interactions between uncoupling protein 2 gene polymorphisms, obesity and alcohol intake on liver function: a large meta-analysed population-based study. Eur J Endocrinol 2015; 173:863-72. [PMID: 26526553 DOI: 10.1530/eje-15-0839] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND AND OBJECTIVE Given the role of uncoupling protein 2 (UCP2) in the accumulation of fat in the hepatocytes and in the enhancement of protective mechanisms in acute ethanol intake, we hypothesised that UCP2 polymorphisms are likely to cause liver disease through their interactions with obesity and alcohol intake. To test this hypothesis, we investigated the interaction between tagging polymorphisms in the UCP2 gene (rs2306819, rs599277 and rs659366), alcohol intake and obesity traits such as BMI and waist circumference (WC) on alanine aminotransferase (ALT) and gamma glutamyl transferase (GGT) in a large meta-analysis of data sets from three populations (n=20 242). DESIGN AND METHODS The study populations included the Northern Finland Birth Cohort 1966 (n=4996), Netherlands Study of Depression and Anxiety (n=1883) and LifeLines Cohort Study (n=13 363). Interactions between the polymorphisms and obesity and alcohol intake on dichotomised ALT and GGT levels were assessed using logistic regression and the likelihood ratio test. RESULTS In the meta-analysis of the three cohorts, none of the three UCP2 polymorphisms were associated with GGT or ALT levels. There was no evidence for interaction between the polymorphisms and alcohol intake on GGT and ALT levels. In contrast, the association of WC and BMI with GGT levels varied by rs659366 genotype (Pinteraction=0.03 and 0.007, respectively; adjusted for age, gender, high alcohol intake, diabetes, hypertension and serum lipid concentrations). CONCLUSION In conclusion, our findings in 20 242 individuals suggest that UCP2 gene polymorphisms may cause liver dysfunction through the interaction with body fat rather than alcohol intake.
Collapse
Affiliation(s)
- Karani S Vimaleswaran
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| | - Alana Cavadino
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| | - Niek Verweij
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia
| | - Ilja M Nolte
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia
| | - Irene Mateo Leach
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia
| | - Juha Auvinen
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| | - Juha Veijola
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| | - Paul Elliott
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia
| | - Brenda W Penninx
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| | - Harold Snieder
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia
| | - Marjo-Riitta Järvelin
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| | - Pim van der Harst
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| | - Robert D Cohen
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia
| | - Barbara J Boucher
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia
| | - Elina Hyppönen
- Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Reading RG6 6AP, UKPopulationPolicy and Practice, UCL Institute of Child Health, London, UKWolfson Institute of Preventive MedicineCentre for Environmental and Preventive Medicine, Queen Mary University of London, London, UK, Departments of CardiologyEpidemiologyUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsUnit of Primary CareOulu University Hospital, Oulu, FinlandFaculty of MedicineCenter for Life Course EpidemiologyDepartment of PsychiatryCenter for Clinical Neuroscience, University of Oulu, Oulu, FinlandDepartment of PsychiatryMedical Research Center, University Hospital of Oulu, Oulu, FinlandDepartment of Epidemiology and BiostatisticsImperial College London, MRC-PHE Centre for Environment and Health, London, UKDepartment of PsychiatryLeiden University Medical Center, Leiden, The NetherlandsDepartment of PsychiatryEMGO Institute of Health and Care Research, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The NetherlandsBiocenter OuluUniversity of Oulu, Oulu, FinlandDepartment of GeneticsUniversity Medical Center Groningen, University of Groningen, Groningen, The NetherlandsICIN - Netherlands Heart InstituteDurrer Center for Cardiogenetic Research, Utrecht, The NetherlandsBarts and The London School of Medicine and DentistryQueen Mary University of London, Blizard Institute, Newark Street, London, UKCentre for Population Health ResearchSchool of Health Science and Sansom Institute of Health Research, University of South Australia, Adelaide, South Australia, AustraliaSouth Australian Health and Medical Research InstituteAdelaide, South Australia, Australia Hugh Sinclair Unit of Human NutritionDepartment of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Whiteknights, PO Box 226, Readin
| |
Collapse
|
203
|
Asada R, Kanemoto S, Matsuhisa K, Hino K, Cui M, Cui X, Kaneko M, Imaizumi K. IRE1α-XBP1 is a novel branch in the transcriptional regulation of Ucp1 in brown adipocytes. Sci Rep 2015; 5:16580. [PMID: 26568450 PMCID: PMC4644985 DOI: 10.1038/srep16580] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/16/2015] [Indexed: 02/08/2023] Open
Abstract
The unfolded protein response (UPR) not only resolves endoplasmic reticulum (ER) stress, but also regulates cellular physiological functions. In this study, we first linked the UPR to the physiological roles of brown adipose tissue (BAT). BAT is one of the tissues that control energy homeostasis in the body. Brown adipocytes are able to dissipate energy in the form of heat owing to their mitochondrial protein, uncoupling protein 1 (UCP1). We found that one of the UPR branches, the IRE1α-XBP1 pathway, was activated during the transcriptional induction of Ucp1. Inhibiting the IRE1α-XBP1 pathway reduced the induction of Ucp1 expression. However, the activation of the IRE1α-XBP1 pathway by ER stress never upregulated Ucp1. On the other hand, the activation of protein kinase A (PKA) induced Ucp1 transcription through the activation of IRE1α-XBP1. The inhibition of PKA abrogated the activation of IRE1α-XBP1 pathway, while the inhibition of a p38 mitogen activated protein kinase (p38 MAPK), which is one of the downstream molecules of PKA, never suppressed the activation of IRE1α-XBP1 pathway. These data indicate that PKA-dependent IRE1α-XBP1 activation is crucial for the transcriptional induction of Ucp1 in brown adipocytes, and they demonstrate a novel, ER stress -independent role of the UPR during thermogenesis.
Collapse
Affiliation(s)
- Rie Asada
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Soshi Kanemoto
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Koji Matsuhisa
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kenta Hino
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Min Cui
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Xiang Cui
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Masayuki Kaneko
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical &Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| |
Collapse
|
204
|
Harmancey R, Haight DL, Watts KA, Taegtmeyer H. Chronic Hyperinsulinemia Causes Selective Insulin Resistance and Down-regulates Uncoupling Protein 3 (UCP3) through the Activation of Sterol Regulatory Element-binding Protein (SREBP)-1 Transcription Factor in the Mouse Heart. J Biol Chem 2015; 290:30947-61. [PMID: 26555260 DOI: 10.1074/jbc.m115.673988] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Indexed: 01/22/2023] Open
Abstract
The risk for heart failure and death after myocardial infarction is abnormally high in diabetic subjects. We and others have shown previously that mitochondrial uncoupling protein 3 (UCP3) improves functional recovery of the rodent heart during reperfusion. Here, we demonstrate that pharmacological induction of hyperinsulinemia in mice down-regulates myocardial UCP3. Decreased UCP3 expression was linked to the development of selective insulin resistance in the heart, characterized by decreased basal activity of Akt but preserved activity of the p44/42 mitogen-activated protein kinase, and overactivation of the sterol regulatory element-binding protein (SREBP)-1-mediated lipogenic program. In cultured myocytes, insulin treatment and SREBP-1 overexpression decreased, whereas SREBP-1 interference increased, peroxisome proliferator-activated receptor-stimulated expression of UCP3. Promoter deletion and site-directed mutagenesis identified three functional sterol regulatory elements in the vicinity of a known complex intronic enhancer. Increased binding of SREBP-1 to this DNA region was confirmed in the heart of hyperinsulinemic mice. In conclusion, we describe a hitherto unknown regulatory mechanism by which insulin inhibits cardiac UCP3 expression through activation of the lipogenic factor SREBP-1. Sustained down-regulation of cardiac UCP3 by hyperinsulinemia may partly explain the poor prognosis of type 2 diabetic patients after myocardial infarction.
Collapse
Affiliation(s)
- Romain Harmancey
- From the Department of Internal Medicine, Division of Cardiology, University of Texas Medical School, University of Texas Health Science Center, Houston, Texas 77030 and the Department of Physiology and Biophysics, Mississippi Center for Obesity Research and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505
| | - Derek L Haight
- From the Department of Internal Medicine, Division of Cardiology, University of Texas Medical School, University of Texas Health Science Center, Houston, Texas 77030 and
| | - Kayla A Watts
- the Department of Physiology and Biophysics, Mississippi Center for Obesity Research and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505
| | - Heinrich Taegtmeyer
- From the Department of Internal Medicine, Division of Cardiology, University of Texas Medical School, University of Texas Health Science Center, Houston, Texas 77030 and
| |
Collapse
|
205
|
Shin HJ, Kwon HK, Lee JH, Gui X, Achek A, Kim JH, Choi S. Doxorubicin-induced necrosis is mediated by poly-(ADP-ribose) polymerase 1 (PARP1) but is independent of p53. Sci Rep 2015; 5:15798. [PMID: 26522181 PMCID: PMC4629133 DOI: 10.1038/srep15798] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022] Open
Abstract
Necrosis, unregulated cell death, is characterized by plasma membrane rupture as well as nuclear and cellular swelling. However, it has recently been reported that necrosis is a regulated form of cell death mediated by poly-(ADP-ribose) polymerase 1 (PARP1). PARP1 is thought to mediate necrosis by inducing DNA damage, although this remains unconfirmed. In this study, we examined the mechanisms of PARP1-mediated necrosis following doxorubicin (DOX)-induced DNA damage in human kidney proximal tubular (HK-2) cells. DOX initiated DNA damage response (DDR) and upregulated PARP1 and p53 expression, resulting in morphological changes similar to those observed during necrosis. Additionally, DOX induced mitochondrial hyper-activation, as evidenced by increased mitochondrial respiration and cytosolic ATP (cATP) production. However, DOX affected mitochondrial mass. DOX-induced DNA damage, cytosolic reactive oxygen species (cROS) generation, and mitochondrial hyper-activation decreased in cells with inhibited PARP1 expression, while generation of nitric oxide (NO) and mitochondrial ROS (mROS) remained unaffected. Moreover, DOX-induced DNA damage, cell cycle changes, and oxidative stress were not affected by p53 inhibition. These findings suggest that DNA damage induced necrosis through a PARP1-dependent and p53-independent pathway.
Collapse
Affiliation(s)
- Hyeon-Jun Shin
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Hyuk-Kwon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Jae-Hyeok Lee
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Xiangai Gui
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Asma Achek
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| |
Collapse
|
206
|
Granger DN, Kvietys PR. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol 2015; 6:524-551. [PMID: 26484802 PMCID: PMC4625011 DOI: 10.1016/j.redox.2015.08.020] [Citation(s) in RCA: 974] [Impact Index Per Article: 97.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/11/2022] Open
Abstract
Reperfusion injury, the paradoxical tissue response that is manifested by blood flow-deprived and oxygen-starved organs following the restoration of blood flow and tissue oxygenation, has been a focus of basic and clinical research for over 4-decades. While a variety of molecular mechanisms have been proposed to explain this phenomenon, excess production of reactive oxygen species (ROS) continues to receive much attention as a critical factor in the genesis of reperfusion injury. As a consequence, considerable effort has been devoted to identifying the dominant cellular and enzymatic sources of excess ROS production following ischemia-reperfusion (I/R). Of the potential ROS sources described to date, xanthine oxidase, NADPH oxidase (Nox), mitochondria, and uncoupled nitric oxide synthase have gained a status as the most likely contributors to reperfusion-induced oxidative stress and represent priority targets for therapeutic intervention against reperfusion-induced organ dysfunction and tissue damage. Although all four enzymatic sources are present in most tissues and are likely to play some role in reperfusion injury, priority and emphasis has been given to specific ROS sources that are enriched in certain tissues, such as xanthine oxidase in the gastrointestinal tract and mitochondria in the metabolically active heart and brain. The possibility that multiple ROS sources contribute to reperfusion injury in most tissues is supported by evidence demonstrating that redox-signaling enables ROS produced by one enzymatic source (e.g., Nox) to activate and enhance ROS production by a second source (e.g., mitochondria). This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current understanding of the four most frequently invoked enzymatic sources of ROS production in post-ischemic tissue. Reperfusion injury is implicated in a variety of human diseases and disorders. Evidence implicating ROS in reperfusion injury continues to grow. Several enzymes are candidate sources of ROS in post-ischemic tissue. Inter-enzymatic ROS-dependent signaling enhances the oxidative stress caused by I/R. .
Collapse
Affiliation(s)
- D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, United States.
| | - Peter R Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
207
|
Normoyle KP, Kim M, Farahvar A, Llano D, Jackson K, Wang H. The emerging neuroprotective role of mitochondrial uncoupling protein-2 in traumatic brain injury. Transl Neurosci 2015; 6:179-186. [PMID: 28123803 PMCID: PMC4936626 DOI: 10.1515/tnsci-2015-0019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/20/2015] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a multifaceted disease with intrinsically complex heterogeneity and remains a significant clinical challenge to manage. TBI model systems have demonstrated many mechanisms that contribute to brain parenchymal cell death, including glutamate and calcium toxicity, oxidative stress, inflammation, and mitochondrial dysfunction. Mitochondria are critically regulated by uncoupling proteins (UCP), which allow protons to leak back into the matrix and thus reduce the mitochondrial membrane potential by dissipating the proton motive force. This uncoupling of oxidative phosphorylation from adenosine triphosphate (ATP) synthesis is potentially critical for protection against cellular injury as a result of TBI and stroke. A greater understanding of the underlying mechanism or mechanisms by which uncoupling protein-2 (UCP2) functions to maintain or optimize mitochondrial function, and the conditions which precipitate the failure of these mechanisms, would inform future research and treatment strategies. We posit that UCP2-mediated function underlies the physiological response to neuronal stress associated with traumatic and ischemic injury and that clinical development of UCP2-targeted treatment would significantly impact these patient populations. With a focus on clinical relevance in TBI, we synthesize current knowledge concerning UCP2 and its potential neuroprotective role and apply this body of knowledge to current and potential treatment modalities.
Collapse
Affiliation(s)
- Kieran P Normoyle
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Miri Kim
- College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Arash Farahvar
- Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, USA
| | - Daniel Llano
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Neurology, Carle Foundation Hospital, Urbana, IL, USA; The Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kevin Jackson
- The Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Thermal Neuroscience Laboratory (TNL), Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Huan Wang
- Department of Neurology, Carle Foundation Hospital, Urbana, IL, USA; Thermal Neuroscience Laboratory (TNL), Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
208
|
Xu Z, Zhang L, Li X, Jiang Z, Sun L, Zhao G, Zhou G, Zhang H, Shang J, Wang T. Mitochondrial fusion/fission process involved in the improvement of catalpol on high glucose-induced hepatic mitochondrial dysfunction. Acta Biochim Biophys Sin (Shanghai) 2015; 47:730-40. [PMID: 26140925 DOI: 10.1093/abbs/gmv061] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/04/2015] [Indexed: 12/26/2022] Open
Abstract
Catalpol, an iridoid glycoside, has been shown to exert hypoglycemic effect by rescuing mitochondrial function, but the detailed mechanism remains unclear yet. In this study, the effect and mechanism of catalpol on the hepatic mitochondria under diabetic conditions were further examined. Oral administration of catalpol significantly reduced the blood glucose, triglyceride, and cholesterol levels in high-fat diet- and streptozotocin-induced diabetic mice. Additionally, catalpol attenuated the decrease in liver mitochondrial ATP content resulting from diabetes. Furthermore, the number of mitochondria possessing a long size was increased in catalpol-treated mice. Interestingly, the catalpol-induced recovery of mitochondrial function was associated with decreased fission protein 1 and dynamin-related protein 1 expression as well as increased mitofusin 1 expression in the liver. In HepG2 cells, catalpol alleviated the decrease of ATP content and mitochondrial membrane potential, and the increase of reactive oxygen species formation induced by high glucose. MitoTracker Green stain shows that the tubular feature of mitochondria was maintained when cells were treated with catalpol. Catalpol also decreased fission protein 1 and dynamin-related protein 1 expression and increased mitofusin 1 expression in HepG2 cells. The present results suggest that catalpol can ameliorate hepatic mitochondrial dysfunction under a diabetic state, and this may be related to its regulation of mitochondrial fusion and fission events.
Collapse
Affiliation(s)
- Zhimeng Xu
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Luyong Zhang
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaojie Li
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Lixin Sun
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Guolin Zhao
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Guohua Zhou
- Qinghai Yangzong Pharmaceutial Co., Ltd, Xining 810003, China
| | - Heran Zhang
- Tianjin Institute of Pharmaceutical Research, Tianjin 300193, China
| | - Jing Shang
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Wang
- Jiangsu Center of Drug Screening, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
209
|
Mitochondrial uncoupling links lipid catabolism to Akt inhibition and resistance to tumorigenesis. Nat Commun 2015; 6:8137. [PMID: 26310111 PMCID: PMC4552083 DOI: 10.1038/ncomms9137] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 07/22/2015] [Indexed: 12/12/2022] Open
Abstract
To support growth, tumour cells reprogramme their metabolism to simultaneously upregulate macromolecular biosynthesis while maintaining energy production. Uncoupling proteins (UCPs) oppose this phenotype by inducing futile mitochondrial respiration that is uncoupled from ATP synthesis, resulting in nutrient wasting. Here using a UCP3 transgene targeted to the basal epidermis, we show that forced mitochondrial uncoupling inhibits skin carcinogenesis by blocking Akt activation. Similarly, Akt activation is markedly inhibited in UCP3 overexpressing primary human keratinocytes. Mechanistic studies reveal that uncoupling increases fatty acid oxidation and membrane phospholipid catabolism, and impairs recruitment of Akt to the plasma membrane. Overexpression of Akt overcomes metabolic regulation by UCP3, rescuing carcinogenesis. These findings demonstrate that mitochondrial uncoupling is an effective strategy to limit proliferation and tumorigenesis through inhibition of Akt, and illuminate a novel mechanism of crosstalk between mitochondrial metabolism and growth signalling.
Collapse
|
210
|
Regulatory networks of non-coding RNAs in brown/beige adipogenesis. Biosci Rep 2015; 35:BSR20150155. [PMID: 26283634 PMCID: PMC4626868 DOI: 10.1042/bsr20150155] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/14/2015] [Indexed: 12/13/2022] Open
Abstract
BAT (brown adipose tissue) is specialized to burn fatty acids for heat generation and energy expenditure to defend against cold and obesity. Accumulating studies have demonstrated that manipulation of BAT activity through various strategies can regulate metabolic homoeostasis and lead to a healthy phenotype. Two classes of ncRNA (non-coding RNA), miRNA and lncRNA (long non-coding RNA), play crucial roles in gene regulation during tissue development and remodelling. In the present review, we summarize recent findings on regulatory role of distinct ncRNAs in brown/beige adipocytes, and discuss how these ncRNA regulatory networks contribute to brown/beige fat development, differentiation and function. We suggest that targeting ncRNAs could be an attractive approach to enhance BAT activity for protecting the body against obesity and its pathological consequences.
Collapse
|
211
|
van Dam AD, Kooijman S, Schilperoort M, Rensen PCN, Boon MR. Regulation of brown fat by AMP-activated protein kinase. Trends Mol Med 2015; 21:571-9. [PMID: 26271143 DOI: 10.1016/j.molmed.2015.07.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/04/2015] [Accepted: 07/07/2015] [Indexed: 12/23/2022]
Abstract
Novel strategies are needed to reduce the obesity epidemic. One promising strategy is activation of brown adipose tissue (BAT), either via the brain or directly, which increases energy expenditure by combustion of fatty acids (FAs) into heat. The enzyme complex AMP-activated protein kinase (AMPK) is crucially involved in energy metabolism and is highly expressed in both brain and BAT, regulating thermogenesis. As a general rule, BAT activity and energy expenditure are increased either by suppression of AMPK activity in the brain, resulting in enhanced sympathetic outflow towards BAT, or by activation of AMPK in BAT. Targeting AMPK may thus hold therapeutic potential for the treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Andrea D van Dam
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, post zone C7Q, PO Box 9600, 2300 RC Leiden, The Netherlands.
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, post zone C7Q, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Maaike Schilperoort
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, post zone C7Q, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, post zone C7Q, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Mariëtte R Boon
- Department of Medicine, Division of Endocrinology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, post zone C7Q, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
212
|
Agil A, Elmahallawy EK, Rodríguez-Ferrer JM, Adem A, Bastaki SM, Al-Abbadi I, Fino Solano YA, Navarro-Alarcón M. Melatonin increases intracellular calcium in the liver, muscle, white adipose tissues and pancreas of diabetic obese rats. Food Funct 2015; 6:2671-8. [PMID: 26134826 DOI: 10.1039/c5fo00590f] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Melatonin, a widespread substance with antioxidant and anti-inflammatory properties, has been found to act as an antidiabetic agent in animal models, regulating the release and action of insulin. However, the molecular bases of this antidiabetic action are unknown, limiting its application in humans. Several studies have recently shown that melatonin can modify calcium (Ca(2+)) in diabetic animals, and Ca(2+) has been reported to be involved in glucose homeostasis. The objective of the present study was to assess whether the antidiabetic effect of chronic melatonin at pharmacological doses is established via Ca(2+) regulation in different tissues in an animal model of obesity-related type 2 diabetes, using Zücker diabetic fatty (ZDF) rats and their lean littermates, Zücker lean (ZL) rats. After the treatments, flame atomic absorption spectrometry was used to determine Ca(2+) levels in the liver, muscle, main types of internal white adipose tissue, subcutaneous lumbar fat, pancreas, brain, and plasma. This study reports for the first time that chronic melatonin administration (10 mg per kg body weight per day for 6 weeks) increases Ca(2+) levels in muscle, liver, different adipose tissues, and pancreas in ZDF rats, although there were no significant changes in their brain or plasma Ca(2+) levels. We propose that this additional peripheral dual action mechanism underlies the improvement in insulin sensitivity and secretion previously documented in samples from the same animals. According to these results, indoleamine may be a potential candidate for the treatment of type 2 diabetes mellitus associated with obesity.
Collapse
Affiliation(s)
- A Agil
- Department of Pharmacology and Neurosciences Institute (CIBM), School of Medicine, University of Granada, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
213
|
Jankovic A, Korac A, Buzadzic B, Otasevic V, Stancic A, Daiber A, Korac B. Redox implications in adipose tissue (dys)function--A new look at old acquaintances. Redox Biol 2015; 6:19-32. [PMID: 26177468 PMCID: PMC4511633 DOI: 10.1016/j.redox.2015.06.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
Obesity is an energy balance disorder associated with dyslipidemia, insulin resistance and diabetes type 2, also summarized with the term metabolic syndrome or syndrome X. Increasing evidence points to “adipocyte dysfunction”, rather than fat mass accretion per se, as the key pathophysiological factor for metabolic complications in obesity. The dysfunctional fat tissue in obesity characterizes a failure to safely store metabolic substrates into existing hypertrophied adipocytes and/or into new preadipocytes recruited for differentiation. In this review we briefly summarize the potential of redox imbalance in fat tissue as an instigator of adipocyte dysfunction in obesity. We reveal the challenge of the adipose redox changes, insights in the regulation of healthy expansion of adipose tissue and its reduction, leading to glucose and lipids overflow. Adipose tissue (AT) buffers nutrient excess determining overall metabolic health. Redox insight in lipid storage and adipogenesis of AT is reviewed. Redox modulation of AT as therapeutic target in obesity/syndrome X is considered.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Aleksandra Korac
- University of Belgrade, Faculty of Biology, Center for Electron Microscopy, Belgrade, Serbia
| | - Biljana Buzadzic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Vesna Otasevic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Ana Stancic
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Andreas Daiber
- 2nd Medical Department, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Bato Korac
- University of Belgrade, Department of Physiology, Institute for Biological Research "Sinisa Stankovic", Bulevar Despota Stefana 142, 11060 Belgrade, Serbia.
| |
Collapse
|
214
|
Lee S, Moon H, Kim G, Cho JH, Dae-Hee L, Ye MB, Park D. Anion Transport or Nucleotide Binding by Ucp2 Is Indispensable for Ucp2-Mediated Efferocytosis. Mol Cells 2015; 38:657-62. [PMID: 26082030 PMCID: PMC4507033 DOI: 10.14348/molcells.2015.0083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/15/2015] [Accepted: 04/16/2015] [Indexed: 11/27/2022] Open
Abstract
Rapid and efficient engulfment of apoptotic cells is an essential property of phagocytes for removal of the large number of apoptotic cells generated in multicellular organisms. To achieve this, phagocytes need to be able to continuously uptake apoptotic cells. It was recently reported that uncoupling protein 2 (Ucp2) promotes engulfment of apoptotic cells by increasing the phagocytic capacity, thereby allowing cells to continuously ingest apoptotic cells. However, the functions of Ucp2, beyond its possible role in dissipating the mitochondrial membrane potential, that contribute to elevation of the phagocytic capacity have not been determined. Here, we report that the anion transfer or nucleotide binding activity of Ucp2, as well as its dissipation of the mitochondrial membrane potential, is necessary for Ucp2-mediated engulfment of apoptotic cells. To study these properties, we generated Ucp2 mutations that affected three different functions of Ucp2, namely, dissipation of the mitochondrial membrane potential, transfer of anions, and binding of purine nucleotides. Mutations of Ucp2 that affected the proton leak did not enhance the engulfment of apoptotic cells. Although anion transfer and nucleotide binding mutations did not affect the mitochondrial membrane potential, they exerted a dominant-negative effect on Ucp2-mediated engulfment. Furthermore, none of our Ucp2 mutations increased the phagocytic capacity. We conclude that dissipation of the proton gradient by Ucp2 is not the only determinant of the phagocytic capacity and that anion transfer or nucleotide binding by Ucp2 is also essential for Ucp2-mediated engulfment of apoptotic cells.
Collapse
Affiliation(s)
- Suho Lee
- School of Life Sciences and Bio Imaging Research Center, Gwangju Institute of Science and Technology, Gwangju 500-712,
Korea
| | - Hyunji Moon
- School of Life Sciences and Bio Imaging Research Center, Gwangju Institute of Science and Technology, Gwangju 500-712,
Korea
- Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750,
Korea
| | - Gayoung Kim
- School of Life Sciences and Bio Imaging Research Center, Gwangju Institute of Science and Technology, Gwangju 500-712,
Korea
- Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750,
Korea
| | - Jeong Hoon Cho
- Department of Biology Education, College of Education, Chosun University, Gwangju 501-759,
Korea
| | - Lee Dae-Hee
- Department of Surgery and Pharmacology and Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA,
USA
| | - Michael B. Ye
- School of Liberal Arts and Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712,
Korea
| | - Daeho Park
- School of Life Sciences and Bio Imaging Research Center, Gwangju Institute of Science and Technology, Gwangju 500-712,
Korea
- Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750,
Korea
| |
Collapse
|
215
|
Tomas E, Stanojevic V, McManus K, Khatri A, Everill P, Bachovchin WW, Habener JF. GLP-1(32-36)amide Pentapeptide Increases Basal Energy Expenditure and Inhibits Weight Gain in Obese Mice. Diabetes 2015; 64:2409-19. [PMID: 25858562 PMCID: PMC4477344 DOI: 10.2337/db14-1708] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/22/2015] [Indexed: 12/31/2022]
Abstract
The prevalence of obesity-related diabetes is increasing worldwide. Here we report the identification of a pentapeptide, GLP-1(32-36)amide (LVKGRamide), derived from the glucoincretin hormone GLP-1, that increases basal energy expenditure and curtails the development of obesity, insulin resistance, diabetes, and hepatic steatosis in diet-induced obese mice. The pentapeptide inhibited weight gain, reduced fat mass without change in energy intake, and increased basal energy expenditure independent of physical activity. Analyses of tissues from peptide-treated mice reveal increased expression of UCP-1 and UCP-3 in brown adipose tissue and increased UCP-3 and inhibition of acetyl-CoA carboxylase in skeletal muscle, findings consistent with increased fatty acid oxidation and thermogenesis. In palmitate-treated C2C12 skeletal myotubes, GLP-1(32-36)amide activated AMPK and inhibited acetyl-CoA carboxylase, suggesting activation of fat metabolism in response to energy depletion. By mass spectroscopy, the pentapeptide is rapidly formed from GLP-1(9-36)amide, the major form of GLP-1 in the circulation of mice. These findings suggest that the reported insulin-like actions of GLP-1 receptor agonists that occur independently of the GLP-1 receptor might be mediated by the pentapeptide, and the previously reported nonapeptide (FIAWLVKGRamide). We propose that by increasing basal energy expenditure, GLP-1(32-36)amide might be a useful treatment for human obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Eva Tomas
- Laboratory of Molecular Endocrinology and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Violeta Stanojevic
- Laboratory of Molecular Endocrinology and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Karen McManus
- Laboratory of Molecular Endocrinology and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ashok Khatri
- Laboratory of Molecular Endocrinology and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Paul Everill
- Department of Biochemistry, Tufts University, Boston, MA
| | | | - Joel F Habener
- Laboratory of Molecular Endocrinology and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
216
|
Uncoupling protein 2 mediates resistance to gemcitabine-induced apoptosis in hepatocellular carcinoma cell lines. Biosci Rep 2015; 35:BSR20150116. [PMID: 26181366 PMCID: PMC4613703 DOI: 10.1042/bsr20150116] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 06/03/2015] [Indexed: 12/24/2022] Open
Abstract
Our results suggest a critical role for mitochondrial uncoupling in gemcitabine (GEM) resistance in hepatocellular carcinoma (HCC) cell lines. Hence, synergistic targeting of uncoupling protein 2 (UCP2) in combination with other chemotherapeutic agents might be more potent in HCC patients. Oxidative stress induction is a common effector pathway for commonly used chemotherapeutic agents like gemcitabine (GEM) in hepatocellular carcinoma (HCC) patients. However, GEM alone or in combination with oxiplatin hardly renders any survival benefits to HCC patients. Mitochondrial uncoupling protein 2 (UCP2) is known to suppress mitochondrial reactive oxygen species (ROS) generation, thus mitigating oxidative stress-induced apoptosis. We demonstrate in the present study, using a panel of HCC cell lines that sensitivity to GEM in HCC well correlate with the endogenous level of UCP2 protein expression. Moreover, ectopic overexpression of UCP2 in a HCC cell line with low endogenous UCP2 expression, HLE, significantly decreased mitochondrial superoxide induction by the anti-cancer drug GEM. Conversely, UCP2 mRNA silencing by RNA interference in HCC cell lines with high endogenous UCP2 expression significantly enhanced GEM-induced mitochondrial superoxide generation and apoptosis. Cumulatively, our results suggest a critical role for mitochondrial uncoupling in GEM resistance in HCC cell lines. Hence, synergistic targeting of UCP2 in combination with other chemotherapeutic agents might be more potent in HCC patients.
Collapse
|
217
|
Hoang T, Kuljanin M, Smith MD, Jelokhani-Niaraki M. A biophysical study on molecular physiology of the uncoupling proteins of the central nervous system. Biosci Rep 2015; 35:e00226. [PMID: 26182433 PMCID: PMC4613710 DOI: 10.1042/bsr20150130] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/04/2015] [Indexed: 01/16/2023] Open
Abstract
Mitochondrial inner membrane uncoupling proteins (UCPs) facilitate transmembrane (TM) proton flux and consequently reduce the membrane potential and ATP production. It has been proposed that the three neuronal human UCPs (UCP2, UCP4 and UCP5) in the central nervous system (CNS) play significant roles in reducing cellular oxidative stress. However, the structure and ion transport mechanism of these proteins remain relatively unexplored. Recently, we reported a novel expression system for obtaining functionally folded UCP1 in bacterial membranes and applied this system to obtain highly pure neuronal UCPs in high yields. In the present study, we report on the structure and function of the three neuronal UCP homologues. Reconstituted neuronal UCPs were dominantly helical in lipid membranes and transported protons in the presence of physiologically-relevant fatty acid (FA) activators. Under similar conditions, all neuronal UCPs also exhibited chloride transport activities that were partially inhibited by FAs. CD, fluorescence and MS measurements and semi-native gel electrophoresis collectively suggest that the reconstituted proteins self-associate in the lipid membranes. Based on SDS titration experiments and other evidence, a general molecular model for the monomeric, dimeric and tetrameric functional forms of UCPs in lipid membranes is proposed. In addition to their shared structural and ion transport features, neuronal UCPs differ in their conformations and proton transport activities (and possibly mechanism) in the presence of different FA activators. The differences in FA-activated UCP-mediated proton transport could serve as an essential factor in understanding and differentiating the physiological roles of UCP homologues in the CNS.
Collapse
Affiliation(s)
- Tuan Hoang
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario, Canada, N2L 3C5 Biophysics Interdepartmental Group, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Miljan Kuljanin
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario, Canada, N2L 3C5
| | - Matthew D Smith
- Department of Biology, Wilfrid Laurier University, Waterloo, Ontario, Canada, N2L 3C5 Biophysics Interdepartmental Group, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Masoud Jelokhani-Niaraki
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario, Canada, N2L 3C5 Biophysics Interdepartmental Group, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| |
Collapse
|
218
|
Gak IA, Radovic SM, Dukic AR, Janjic MM, Stojkov-Mimic NJ, Kostic TS, Andric SA. Stress triggers mitochondrial biogenesis to preserve steroidogenesis in Leydig cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2217-27. [PMID: 26036344 DOI: 10.1016/j.bbamcr.2015.05.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/25/2015] [Accepted: 05/29/2015] [Indexed: 12/12/2022]
Abstract
Adaptability to stress is a fundamental prerequisite for survival. Mitochondria are a key component of the stress response in all cells. For steroid-hormones-producing cells, including also Leydig cells of testes, the mitochondria are a key control point for the steroid biosynthesis and regulation. However, the mitochondrial biogenesis in steroidogenic cells has never been explored. Here we show that increased mitochondrial biogenesis is the adaptive response of testosterone-producing Leydig cells from stressed rats. All markers of mitochondrial biogenesis together with transcription factors and related kinases are up-regulated in Leydig cells from rats exposed to repeated psychophysical stress. This is followed with increased mitochondrial mass. The expression of PGC1, master regulator of mitochondrial biogenesis and integrator of environmental signals, is stimulated by cAMP-PRKA, cGMP, and β-adrenergic receptors. Accordingly, stress-triggered mitochondrial biogenesis represents an adaptive mechanism and does not only correlate with but also is an essential for testosterone production, being both events depend on the same regulators. Here we propose that all events induced by acute stress, the most common stress in human society, provoke adaptive response of testosterone-producing Leydig cells and activate PGC1, a protein required to make new mitochondria but also protector against the oxidative damage. Given the importance of mitochondria for steroid hormones production and stress response, as well as the role of steroid hormones in stress response and metabolic syndrome, we anticipate our result to be a starting point for more investigations since stress is a constant factor in life and has become one of the most significant health problems in modern societies.
Collapse
Affiliation(s)
- Igor A Gak
- Laboratory for Reproductive Endocrinology and Signaling (LaRES), Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Sq. 2, 21000 Novi Sad, Serbia
| | - Sava M Radovic
- Laboratory for Reproductive Endocrinology and Signaling (LaRES), Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Sq. 2, 21000 Novi Sad, Serbia
| | - Aleksandra R Dukic
- Laboratory for Reproductive Endocrinology and Signaling (LaRES), Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Sq. 2, 21000 Novi Sad, Serbia
| | - Marija M Janjic
- Laboratory for Reproductive Endocrinology and Signaling (LaRES), Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Sq. 2, 21000 Novi Sad, Serbia
| | - Natasa J Stojkov-Mimic
- Laboratory for Reproductive Endocrinology and Signaling (LaRES), Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Sq. 2, 21000 Novi Sad, Serbia
| | - Tatjana S Kostic
- Laboratory for Reproductive Endocrinology and Signaling (LaRES), Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Sq. 2, 21000 Novi Sad, Serbia
| | - Silvana A Andric
- Laboratory for Reproductive Endocrinology and Signaling (LaRES), Faculty of Sciences, University of Novi Sad, Dositeja Obradovica Sq. 2, 21000 Novi Sad, Serbia.
| |
Collapse
|
219
|
Nishikawa K, Iwaya K, Kinoshita M, Fujiwara Y, Akao M, Sonoda M, Thiruppathi S, Suzuki T, Hiroi S, Seki S, Sakamoto T. Resveratrol increases CD68⁺ Kupffer cells colocalized with adipose differentiation-related protein and ameliorates high-fat-diet-induced fatty liver in mice. Mol Nutr Food Res 2015; 59:1155-70. [PMID: 25677089 DOI: 10.1002/mnfr.201400564] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 12/27/2014] [Accepted: 01/20/2015] [Indexed: 12/12/2022]
Abstract
SCOPE Resveratrol reportedly improves fatty liver. This study purposed to elucidate the effect of resveratrol on fatty liver in mice fed a high-fat (HF) diet, and to investigate the role of liver macrophages (Kupffer cells). METHODS AND RESULTS C57BL/6 mice were divided into three groups, receiving either a control diet, HF diet (50% fat), or HF supplemented with 0.2% resveratrol (HF + res) diet, for 8 weeks. Compared with the HF group, the HF + res group exhibited markedly attenuated fatty liver, and reduced lipid droplets (LDs) in hepatocytes. Proteomic analysis demonstrated that the most downregulated protein in the livers of the HF + res group was adipose differentiation-related protein (ADFP), which is a major constituent of LDs and reflects lipid accumulation in cells. The HF + res group exhibited greatly increased numbers of CD68(+) Kupffer cells with phagocytic activity. Immunohistochemistry showed that several CD68(+) Kupffer cells were colocalized with ADFP immunoreaction in the HF + res group. Additionally, the HF + res group demonstrated markedly decreased TNF-alpha production, which confirmed by both liver mononuclear cells stimulated by LPS in vitro and in situ hybridization analysis, compared with the HF group. CONCLUSION Resveratrol ameliorated fatty liver and increased CD68-positive Kupffer cells with downregulating ADFP expression.
Collapse
Affiliation(s)
| | - Keiichi Iwaya
- Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Saitama, Japan
| | - Yoko Fujiwara
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Mai Akao
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Mariko Sonoda
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Suresh Thiruppathi
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, Japan
| | - Takayoshi Suzuki
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, Japan
| | - Sadayuki Hiroi
- Department of Laboratory Medicine, National Defense Medical College, Saitama, Japan
| | - Shuhji Seki
- Department of Immunology and Microbiology, National Defense Medical College, Saitama, Japan
| | - Toshihisa Sakamoto
- Department of Traumatology and Critical Care Medicine, National Defense Medical College, Saitama, Japan
| |
Collapse
|
220
|
Gu Q, Zhao L, Ma YP, Liu JD. Contribution of mitochondrial function to exercise-induced attenuation of renal dysfunction in spontaneously hypertensive rats. Mol Cell Biochem 2015; 406:217-25. [PMID: 25963667 DOI: 10.1007/s11010-015-2439-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/06/2015] [Indexed: 02/01/2023]
Abstract
It is well known that exercise training exhibits renal protective effects in animal models of hypertension and chronic renal failure. However, the mechanisms regulating these effects of exercise training remain unclear. This study aimed to investigate the role of mitochondrial function in exercise-induced attenuation of renal injury in spontaneously hypertensive rats (SHR). The adult male SHR and age-matched normotensive Wistar-Kyoto rats (WKY) were given moderate-intensity exercise for 12 weeks or treated with MitoQ10 for 8 weeks. In this work, exercise training in SHR reduced blood pressure, and effectively attenuated renal dysfunction, marked by reduced creatinine excretion, albuminuria, blood urea nitrogen, and glomerular sclerosis. Exercise training in SHR reduced MDA levels in plasma and kidneys and suppressed formation of 3-nitrotyrosine in kidneys. Exercise training suppressed mitochondrial ROS and [Formula: see text] formation, enhanced ATP formation, reduced mitochondrial swelling, and restored electron transport chain enzyme activity in kidneys of SHR. Furthermore, exercise training upregulated protein expression of uncoupling protein 2 and manganese superoxide dismutase in kidneys of SHR. In addition, treatment with mitochondria-targeted antioxidant MitoQ10 exhibited similar renal protective effects in SHR. In conclusion, chronic aerobic exercise training preserved mitochondrial function and abated oxidative stress in the kidneys of SHR, which may in part explain the protective effect of exercise on renal function and structure in hypertensive individuals.
Collapse
Affiliation(s)
- Qi Gu
- School of Physical Education, Xi'an Technological University, 4 Jinhua Road, Xi'an, 710032, Shaanxi Province, China,
| | | | | | | |
Collapse
|
221
|
Lopimune-induced mitochondrial toxicity is attenuated by increased uncoupling protein-2 level in treated mouse hepatocytes. Biochem J 2015; 468:401-7. [PMID: 26173235 DOI: 10.1042/bj20150195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/13/2015] [Indexed: 02/01/2023]
Abstract
Although the protease inhibitor (PI) Lopimune has proven to be effective, no studies have examined the side effects of Lopimune on mitochondrial bioenergetics in hepatocytes. The objective of the present study is to evaluate mitochondrial respiration, production of reactive oxygen species (ROS) and expression of uncoupling protein-2 (UCP2) in mouse hepatocytes following Lopimune administration. Mitochondria were extracted from mouse liver using differential centrifugation and hepatocytes were isolated by the collagenase perfusion procedure. Mitochondrial respiration was measured using a Rank Brothers oxygen electrode. ROS production in hepatocytes was monitored by flow cytometry using a 2',7'-dichlorofluorescin diacetate probe and UCP2 protein expression was detected by Western blotting. We found that Lopimune induced a significant decrease of approximately 30% in the respiratory control ratio (RCR) starting from day 4 until day 9 of treatment. This decrease was due to an increase in state 4 respiration, reflecting an increase in mitochondrial proton leak. State 2 and state 3 respirations were not affected. Moreover, ROS production significantly increased by about 2-fold after day 1 of treatment and decreased after day 3, returning to the resting level on day 5. Interestingly, UCP2 which is absent from control hepatocytes, was expressed starting from day 4 of treatment. Our findings indicate that Lopimune-induced proton leak, mediated by UCP2, may represent a response to inhibit the production of ROS as a negative feedback regulatory mechanism. These results imply a potential involvement of UCP2 in the regulation of oxidative stress and add new insights into the understanding of mitochondrial toxicity induced by PIs.
Collapse
|
222
|
Metabolic signatures of renal cell carcinoma. Biochem Biophys Res Commun 2015; 460:938-43. [PMID: 25839656 DOI: 10.1016/j.bbrc.2015.03.130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 03/19/2015] [Indexed: 11/21/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by the constitutive up-regulation of the hypoxia inducible factor-1. One of its target enzymes, pyruvate dehydrogenase (PDH) kinase 1 (PDHK1) showed increased protein expression in tumor as compared to patient-matched normal tissues. PDHK1 phosphorylated and inhibited PDH whose enzymatic activity was severely diminished, depriving the TCA cycle of acetylCoA. We and others have shown a decrease in the protein expressions of all respiratory complexes alluding to a compromise in oxidative phosphorylation (OXPHOS). On the contrary, we found that key parameters of OXPHOS, namely ATP biosynthesis and membrane potential were consistently measurable in mitochondria isolated from ccRCC tumor tissues. Interestingly, an endogenous mitochondrial membrane potential (MMP) was evident when ADP was added to mitochondria isolated from ccRCC but not in normal tissues. In addition, the MMP elicited in the presence of ADP by respiratory substrates namely malate/glutamate, succinate, α-ketoglutarate and isocitrate was invariably higher in ccRCC. Two additional hallmarks of ccRCC include a loss of uncoupling protein (UCP)-2 and an increase in UCP-3. Based on our data, we proposed that inhibition of UCP3 by ADP could contribute to the endogenous MMP observed in ccRCC and other cancer cells.
Collapse
|
223
|
Thompson JW, Narayanan SV, Koronowski KB, Morris-Blanco K, Dave KR, Perez-Pinzon MA. Signaling pathways leading to ischemic mitochondrial neuroprotection. J Bioenerg Biomembr 2015; 47:101-10. [PMID: 25262285 PMCID: PMC4861652 DOI: 10.1007/s10863-014-9574-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/20/2014] [Indexed: 12/11/2022]
Abstract
There is extensive evidence that ischemic/reperfusion mediated mitochondrial dysfunction is a major contributor to ischemic damage. However data also indicates that mild ischemic stress induces mitochondrial dependent activation of ischemic preconditioning. Ischemic preconditioning is a neuroprotective mechanism which is activated upon a brief sub-injurious ischemic exposure and is sufficient to provide protection against a subsequent lethal ischemic insult. Current research demonstrates that mitochondria are not only the inducers of but are also an important target of ischemic preconditioning mediated protection. Numerous proteins and signaling pathways are activated by ischemic preconditioning which protect the mitochondria against ischemic damage. In this review we examine some of the proteins activated by ischemic precondition which counteracts the deleterious effects of ischemia/reperfusion thereby maintaining normal mitochondrial activity and lead to ischemic tolerance.
Collapse
Affiliation(s)
- John W Thompson
- Cerebral Vascular Disease Research Laboratories, Department of Neurology and Neuroscience Program, Miller School of Medicine, University of Miami, P.O. Box 016960, Miami, FL, 33136, USA
| | | | | | | | | | | |
Collapse
|
224
|
Wen ZY, Liang XF, He S, Li L, Shen D, Tao YX. Molecular cloning and tissue expression of uncoupling protein 1, 2 and 3 genes in Chinese perch (Siniperca chuatsi). Comp Biochem Physiol B Biochem Mol Biol 2015; 185:24-33. [PMID: 25829150 DOI: 10.1016/j.cbpb.2015.03.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/17/2015] [Accepted: 03/22/2015] [Indexed: 01/07/2023]
Abstract
Uncoupling proteins (UCPs) are mitochondrial anion carrier proteins, which play important roles in several physiological processes, including thermogenesis, reactive oxygen species generation, growth, lipid metabolism and insulin secretion. Although the roles of UCPs are well understood in mammals, little is known in fish. To investigate the thermogenesis roles in Chinese perch (Siniperca chuatsi), we cloned the UCP1, 2 and 3. The UCP1 consisted of six exons and five introns, and the UCP2 consisted of eight exons and seven introns. The UCP1 was primarily expressed in liver, UCP2 was ubiquitously expressed, and UCP3 was primarily expressed in muscle. The mRNA levels of UCP1 and UCP2 in liver, and UCP3 in muscle were significantly increased after prolonged cold exposure, but did not change after prolonged heat exposure, suggesting that Chinese perch might have a mechanism of response to cold environment, but not to hot environment. The intestinal UCP1 mRNA level was significantly up-regulated after prolonged heat exposure, while the UCP2 mRNA level was significantly up-regulated after prolonged cold exposure, suggesting that the two paralogs might play different roles in intestine of Chinese perch. In addition, the phylogenetic analysis could shed new light on the evolutionary diversification of UCP gene family.
Collapse
Affiliation(s)
- Zheng-Yong Wen
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China
| | - Xu-Fang Liang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China.
| | - Shan He
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China
| | - Ling Li
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China
| | - Dan Shen
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849-5519, USA
| |
Collapse
|
225
|
Hoang T, Matovic T, Parker J, Smith MD, Jelokhani-Niaraki M. Role of positively charged residues of the second transmembrane domain in the ion transport activity and conformation of human uncoupling protein-2. Biochemistry 2015; 54:2303-13. [PMID: 25789405 DOI: 10.1021/acs.biochem.5b00177] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Residing at the inner mitochondrial membrane, uncoupling protein-2 (UCP2) mediates proton transport from the intermembrane space (IMS) to the mitochondrial matrix and consequently reduces the rate of ATP synthesis in the mitochondria. The ubiquitous expression of UCP2 in humans can be attributed to the protein's multiple physiological roles in tissues, including its involvement in protective mechanisms against oxidative stress, as well as glucose and lipid metabolisms. Currently, the structural properties and ion transport mechanism of UCP2 and other UCP homologues remain poorly understood. UCP2-mediated proton transport is activated by fatty acids and inhibited by di- and triphosphate purine nucleotides. UCP2 also transports chloride and some other small anions. Identification of key amino acid residues of UCP2 in its ion transport pathway can shed light on the protein's ion transport function. On the basis of our previous studies, the second transmembrane helix segment (TM2) of UCP2 exhibited chloride channel activity. In addition, it was suggested that the positively charged residues on TM2 domains of UCPs 1 and 2 were important for their chloride transport activity. On this basis, to further understand the role of these positively charged residues on the ion transport activity of UCP2, we recombinantly expressed four TM2 mutants: R76Q, R88Q, R96Q, and K104Q. The wild type UCP2 and its mutants were purified and reconstituted into liposomes, and their conformation and ion (proton and chloride) transport activity were studied. TM2 Arg residues at the matrix interface of UCP2 proved to be crucial for the protein's anion transport function, and their absence resulted in highly diminished Cl(-) transport rates. On the other hand, the two other positively charged residues of TM2, located at the UCP2-IMS interface, could participate in the salt-bridge formation in the protein and promote the interhelical tight packing in the UCP2. Absence of these residues did not influence Cl(-) transport rates, but disturbed the dense packing in UCP2 and resulted in higher UCP2-mediated proton transport rates in the presence of long chain fatty acids. Overall, the outcome of this study provides a deeper and more detailed molecular image of UCP2's ion transport mechanism.
Collapse
Affiliation(s)
- Tuan Hoang
- §Biophysics Interdepartmental Group, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | | | | - Matthew D Smith
- §Biophysics Interdepartmental Group, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | |
Collapse
|
226
|
Li J, Zhao WG, Shen ZF, Yuan T, Liu SN, Liu Q, Fu Y, Sun W. Comparative proteome analysis of brown adipose tissue in obese C57BL/6J mice using iTRAQ-coupled 2D LC-MS/MS. PLoS One 2015; 10:e0119350. [PMID: 25747866 PMCID: PMC4352050 DOI: 10.1371/journal.pone.0119350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/12/2015] [Indexed: 12/14/2022] Open
Abstract
High-fat diet (HFD) leads to the development of obesity accompanied by insulin resistance, which increases the risk of type 2 diabetes mellitus and cardiovascular disease. Brown adipose tissue (BAT) plays an essential role in energy metabolism, thus it will give us promising treatment targets through elucidating underlying mechanisms of BAT in obesity. In this study, female C57BL/6J mice were fed HFD or normal diet (ND) for 22 weeks. Hyperinsulinemic-euglycemic clamp was performed to evaluate insulin sensitivity, which was independently correlated with obesity. Using isobaric tag for relative and absolute quantification (iTRAQ) coupled with 2D LC-MS/MS, we quantitated 3048 proteins in BAT. As compared HFD with ND, we obtained 727 differentially expressed proteins. Functional analysis found that those proteins were mainly assigned to the pathway of mitochondrial function. In this pathway, carnitine O-palmitoyltransferase 2 (CPT2), uncoupling protein 1 (UCP1) and apoptosis-inducing factor 1 (AIF1) were up-regulated significantly by HFD, and they were confirmed by western blotting. The results indicated that HFD might induce the apoptosis of brown adipocytes via the up-regulated AIF1. Meanwhile, HFD also stimulated fatty acid β-oxidation and raised compensatory energy consuming through the increases of CPT2 and UCP1, respectively. However, the apoptosis of brown adipocytes might weaken the compensatory energy expenditure, and finally contribute to overweight/obesity. So, preventing the apoptosis of brown adipocytes may be the key target to treat obesity.
Collapse
Affiliation(s)
- Juan Li
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei-Gang Zhao
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- * E-mail: (WGZ); (WS)
| | - Zhu-Fang Shen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Yuan
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Shuai-Nan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
| | - Quan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Fu
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences/School of Basic Medicine, Peking Union Medical College, Beijing, China
- * E-mail: (WGZ); (WS)
| |
Collapse
|
227
|
Busiello RA, Savarese S, Lombardi A. Mitochondrial uncoupling proteins and energy metabolism. Front Physiol 2015; 6:36. [PMID: 25713540 PMCID: PMC4322621 DOI: 10.3389/fphys.2015.00036] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/23/2015] [Indexed: 12/17/2022] Open
Abstract
Understanding the metabolic factors that contribute to energy metabolism (EM) is critical for the development of new treatments for obesity and related diseases. Mitochondrial oxidative phosphorylation is not perfectly coupled to ATP synthesis, and the process of proton-leak plays a crucial role. Proton-leak accounts for a significant part of the resting metabolic rate (RMR) and therefore enhancement of this process represents a potential target for obesity treatment. Since their discovery, uncoupling proteins have stimulated great interest due to their involvement in mitochondrial-inducible proton-leak. Despite the widely accepted uncoupling/thermogenic effect of uncoupling protein one (UCP1), which was the first in this family to be discovered, the reactions catalyzed by its homolog UCP3 and the physiological role remain under debate. This review provides an overview of the role played by UCP1 and UCP3 in mitochondrial uncoupling/functionality as well as EM and suggests that they are a potential therapeutic target for treating obesity and its related diseases such as type II diabetes mellitus.
Collapse
Affiliation(s)
- Rosa A Busiello
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio Benevento, Italy
| | - Sabrina Savarese
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi di Napoli Caserta, Italy
| | - Assunta Lombardi
- Dipartimento di Biologia, Università degli Studi di Napoli Napoli, Italy
| |
Collapse
|
228
|
Uncoupling lipid metabolism from inflammation through fatty acid binding protein-dependent expression of UCP2. Mol Cell Biol 2015; 35:1055-65. [PMID: 25582199 DOI: 10.1128/mcb.01122-14] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Chronic inflammation in obese adipose tissue is linked to endoplasmic reticulum (ER) stress and systemic insulin resistance. Targeted deletion of the murine fatty acid binding protein (FABP4/aP2) uncouples obesity from inflammation although the mechanism underlying this finding has remained enigmatic. Here, we show that inhibition or deletion of FABP4/aP2 in macrophages results in increased intracellular free fatty acids (FFAs) and elevated expression of uncoupling protein 2 (UCP2) without concomitant increases in UCP1 or UCP3. Silencing of UCP2 mRNA in FABP4/aP2-deficient macrophages negated the protective effect of FABP loss and increased ER stress in response to palmitate or lipopolysaccharide (LPS). Pharmacologic inhibition of FABP4/aP2 with the FABP inhibitor HTS01037 also upregulated UCP2 and reduced expression of BiP, CHOP, and XBP-1s. Expression of native FABP4/aP2 (but not the non-fatty acid binding mutant R126Q) into FABP4/aP2 null cells reduced UCP2 expression, suggesting that the FABP-FFA equilibrium controls UCP2 expression. FABP4/aP2-deficient macrophages are resistant to LPS-induced mitochondrial dysfunction and exhibit decreased mitochondrial protein carbonylation and UCP2-dependent reduction in intracellular reactive oxygen species. These data demonstrate that FABP4/aP2 directly regulates intracellular FFA levels and indirectly controls macrophage inflammation and ER stress by regulating the expression of UCP2.
Collapse
|
229
|
Superresolution microscopy reveals spatial separation of UCP4 and F0F1-ATP synthase in neuronal mitochondria. Proc Natl Acad Sci U S A 2014; 112:130-5. [PMID: 25535394 DOI: 10.1073/pnas.1415261112] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Because different proteins compete for the proton gradient across the inner mitochondrial membrane, an efficient mechanism is required for allocation of associated chemical potential to the distinct demands, such as ATP production, thermogenesis, regulation of reactive oxygen species (ROS), etc. Here, we used the superresolution technique dSTORM (direct stochastic optical reconstruction microscopy) to visualize several mitochondrial proteins in primary mouse neurons and test the hypothesis that uncoupling protein 4 (UCP4) and F0F1-ATP synthase are spatially separated to eliminate competition for the proton motive force. We found that UCP4, F0F1-ATP synthase, and the mitochondrial marker voltage-dependent anion channel (VDAC) have various expression levels in different mitochondria, supporting the hypothesis of mitochondrial heterogeneity. Our experimental results further revealed that UCP4 is preferentially localized in close vicinity to VDAC, presumably at the inner boundary membrane, whereas F0F1-ATP synthase is more centrally located at the cristae membrane. The data suggest that UCP4 cannot compete for protons because of its spatial separation from both the proton pumps and the ATP synthase. Thus, mitochondrial morphology precludes UCP4 from acting as an uncoupler of oxidative phosphorylation but is consistent with the view that UCP4 may dissipate the excessive proton gradient, which is usually associated with ROS production.
Collapse
|
230
|
Chen SF, Rao LY, Wei TZ, Xu MG, Dong ZL. Expression patterns of sarcomeric α-actin, α-actinin and UCP2 in the myocardium of Kunming mice after exposure to c-terminal polypeptide of cardiotrophin-1. ACTA ACUST UNITED AC 2014; 34:796-800. [PMID: 25480572 DOI: 10.1007/s11596-014-1355-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/01/2014] [Indexed: 11/24/2022]
Abstract
Cardiotrophin-1 (CT-1) activates a distinct form of cardiac muscle cell hypertrophy in which the sarcomeric units are assembled in series. The aim of the study was to determine the expression pattern of sarcomeric contractile protein α-actin, specialized cytoskeletal protein α-actinin and mitochondrial uncoupling protein-2 (UCP2) in myocardial remodeling induced by chronic exposure to CT-1. Kunming mice were intraperitoneally injected with carboxy-terminal polypeptide (CP) of CT-1 (CT-1-CP, 500 μg·kg(-1)· day(-1)) for 1, 2, 3 and 4 week (s), respectively (4 groups obtained according to the injection time, n=10 each, with 5 males and 5 females in each group). Those injected with physiological saline for 4 weeks served as controls (n=10, with 5 males and 5 females). The heart tissues of mice were harvested at 1, 2, 3 or 4 week (s). Immunohistochemistry (IHC) and Western blotting (WB) were used to detect the distribution and expression of sarcomeric α-actin, α-actinin and mitochondrial UCP2 in myocardial tissues. IHC showed that α-actin was mainly distributed around the nuclei of cardiomyocytes, α-actinin concentrated around the striae and UCP2 scattered rather evenly in the plasma. The expression of α-actin was slightly greater than that of α-actinin and UCP2 in the control group (IHC: χ(2)=6.125; WB: F=0.249, P>0.05) and it gradually decreased after exposure to CT-1-CP. There was no significant difference in the expression of α-actin between the control group and the CT-1-CP-treated groups (χ (2)=7.386, P>0.05). But Western blotting revealed significant difference in the expression of α-actin between the control group and the 4-week CT-1-CP-treated group (F=2.912; q=4.203, P<0.05). Moreover, it was found that the expression of α-actinin increased stepwise with the exposure time in CT-1-CP-treated groups and differed significantly between CT-1-CP-treated groups and the control group (ICH: χ (2)=21.977; WB: F=50.388; P<0.01). The expression of UCP2 was initially increased (WB: control group vs. 1- or 2-week group, q values: 5.603 and 9.995, respectively, P<0.01) and then decreased (WB: control group vs. 3-week group, q=4.742, P<0.01; control group vs. 4-week group, q=0.558, P>0.05). It was suggested that long-term exposure to CT-1-CP could lead to the alteration in the expression of sarcomeric α-actin, α-actinin and mitochondrial UCP2. The different expressions of sarcomeric structure proteins and mitochondrial UCP2 may be involved in myocardial remodeling.
Collapse
Affiliation(s)
- Shu-Fen Chen
- Central Clinical Laboratory, Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Li-Ya Rao
- Department of Cardiology Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao-Zhi Wei
- Department of Cardiology Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min-Guang Xu
- Department of Physiology, Hainan Medical University, Haikou, 571101, China
| | - Zhan-Ling Dong
- Department of Physiology, Hainan Medical University, Haikou, 571101, China
| |
Collapse
|
231
|
Abstract
An alteration of energy balance is the immediate cause of the so-called cachexia. Although alterations of energy intake are often associated with cachexia, it has lately became clear that an increased energy expenditure is the main cause of wasting associated with different types of pathological conditions, such as cancer, infections or chronic heart failure among others. Different types of molecular mechanisms contribute to energy expenditure and, therefore, involuntary body weight loss; among them, adenosine triphosphate (ATP) consumption by sarcoplasmic reticulum Ca(2+) pumps could represent a key mechanism. In other cases, an increase in energy inefficiency will further contribute to energy imbalance.
Collapse
|
232
|
Abstract
The ability of an organism to convert organic molecules from the environment into energy is essential for the development of cellular structures, cell differentiation and growth. Mitochondria have a fundamental role in regulating metabolic pathways, and tight control of mitochondrial functions and dynamics is critical to maintaining adequate energy balance. In complex organisms, such as mammals, it is also essential that the metabolic demands of various tissues are coordinated to ensure that the energy needs of the whole body are effectively met. Within the arcuate nucleus of the hypothalamus, the NPY-AgRP and POMC neurons have a crucial role in orchestrating the regulation of hunger and satiety. Emerging findings from animal studies have revealed an important function for mitochondrial dynamics within these two neuronal populations, which facilitates the correct adaptive responses of the whole body to changes in the metabolic milieu. The main proteins implicated in these studies are the mitofusins, Mfn1 and Mfn2, which are regulators of mitochondrial dynamics. In this Review, we provide an overview of the mechanisms by which mitochondria are involved in the central regulation of energy balance and discuss the implications of mitochondrial dysfunction for metabolic disorders.
Collapse
Affiliation(s)
- Carole M Nasrallah
- Program in Integrative Cell Signalling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, SHM L-200, PO Box 208074, New Haven, CT 06520-8074, USA
| | - Tamas L Horvath
- Program in Integrative Cell Signalling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, SHM L-200, PO Box 208074, New Haven, CT 06520-8074, USA
| |
Collapse
|
233
|
Abstract
Obesity and related diseases are a major cause of human morbidity and mortality and constitute a substantial economic burden for society. Effective treatment regimens are scarce, and new therapeutic targets are needed. Brown adipose tissue, an energy-expending tissue that produces heat, represents a potential therapeutic target. Its presence is associated with low body mass index, low total adipose tissue content and a lower risk of type 2 diabetes mellitus. Knowledge about the development and function of thermogenic adipocytes in brown adipose tissue has increased substantially in the last decade. Important transcriptional regulators have been identified, and hormones able to modulate the thermogenic capacity of the tissue have been recognized. Intriguingly, it is now clear that humans, like rodents, possess two types of thermogenic adipocytes: the classical brown adipocytes found in the interscapular brown adipose organ and the so-called beige adipocytes primarily found in subcutaneous white adipose tissue after adrenergic stimulation. The presence of two distinct types of energy-expending adipocytes in humans is conceptually important because these cells might be stimulated and recruited by different signals, raising the possibility that they might be separate potential targets for therapeutic intervention. In this review, we will discuss important features of the energy-expending brown adipose tissue and highlight those that may serve as potential targets for pharmacological intervention aimed at expanding the tissue and/or enhancing its function to counteract obesity.
Collapse
Affiliation(s)
- M E Lidell
- Department of Medical and Clinical Genetics, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | |
Collapse
|
234
|
Geigenberger P, Fernie AR. Metabolic control of redox and redox control of metabolism in plants. Antioxid Redox Signal 2014; 21:1389-421. [PMID: 24960279 PMCID: PMC4158967 DOI: 10.1089/ars.2014.6018] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Reduction-oxidation (Redox) status operates as a major integrator of subcellular and extracellular metabolism and is simultaneously itself regulated by metabolic processes. Redox status not only dominates cellular metabolism due to the prominence of NAD(H) and NADP(H) couples in myriad metabolic reactions but also acts as an effective signal that informs the cell of the prevailing environmental conditions. After relay of this information, the cell is able to appropriately respond via a range of mechanisms, including directly affecting cellular functioning and reprogramming nuclear gene expression. RECENT ADVANCES The facile accession of Arabidopsis knockout mutants alongside the adoption of broad-scale post-genomic approaches, which are able to provide transcriptomic-, proteomic-, and metabolomic-level information alongside traditional biochemical and emerging cell biological techniques, has dramatically advanced our understanding of redox status control. This review summarizes redox status control of metabolism and the metabolic control of redox status at both cellular and subcellular levels. CRITICAL ISSUES It is becoming apparent that plastid, mitochondria, and peroxisome functions influence a wide range of processes outside of the organelles themselves. While knowledge of the network of metabolic pathways and their intraorganellar redox status regulation has increased in the last years, little is known about the interorganellar redox signals coordinating these networks. A current challenge is, therefore, synthesizing our knowledge and planning experiments that tackle redox status regulation at both inter- and intracellular levels. FUTURE DIRECTIONS Emerging tools are enabling ever-increasing spatiotemporal resolution of metabolism and imaging of redox status components. Broader application of these tools will likely greatly enhance our understanding of the interplay of redox status and metabolism as well as elucidating and characterizing signaling features thereof. We propose that such information will enable us to dissect the regulatory hierarchies that mediate the strict coupling of metabolism and redox status which, ultimately, determine plant growth and development.
Collapse
Affiliation(s)
- Peter Geigenberger
- 1 Department of Biology I, Ludwig Maximilian University Munich , Planegg-Martinsried, Germany
| | | |
Collapse
|
235
|
Perspectives on mitochondrial uncoupling proteins-mediated neuroprotection. J Bioenerg Biomembr 2014; 47:119-31. [PMID: 25217852 DOI: 10.1007/s10863-014-9580-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
The integrity of mitochondrial function is essential to cell life. It follows that disturbances of mitochondrial function will lead to disruption of cell function, expressed as disease or even death. Considering that neuronal uncoupling proteins (UCPs) decrease reactive oxygen species (ROS) production at the expense of energy production, it is important to understand the underlying mechanisms by which UCPs control the balance between the production of adenosine triphosphate (ATP) and ROS in the context of normal physiological activity and in pathological conditions. Here we review the current understanding of neuronal UCPs-mediated respiratory uncoupling process by performing a survey in their physiology and regulation. The latest findings regarding neuronal UCPs physiological roles and their involvement and interest as potential targets for therapeutic intervention in brain diseases will also be exploited.
Collapse
|
236
|
Berardi MJ, Chou JJ. Fatty acid flippase activity of UCP2 is essential for its proton transport in mitochondria. Cell Metab 2014; 20:541-52. [PMID: 25127353 PMCID: PMC4156518 DOI: 10.1016/j.cmet.2014.07.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/06/2014] [Accepted: 07/02/2014] [Indexed: 01/11/2023]
Abstract
Modulation of cellular energy expenditure is fundamental to normal and pathological cell growth and differentiation. Mitochondria stores energy as a proton gradient across their inner membrane. Uncoupling proteins (UCPs) can dissipate the gradient to produce heat or regulate metabolite fluxes. UCP-mediated proton currents require fatty acids (FAs) and are blocked by nucleotides, but the molecular basis of these processes is unknown. We find, by nuclear magnetic resonance and functional mutagenesis, that UCP2 can bind FAs laterally through its peripheral site, and this intramembrane molecular recognition is essential for UCP2-catalyzed FA flipping across the membrane, which in turn is essential for proton translocation. The antagonist GDP binds inside the UCP2 cavity and perturbs its conformation, which can displace FA from the peripheral site as a mean of inhibiting proton currents. Our data provide a biophysical perspective of the intricate interplay of UCPs, FA, and nucleotides in determining proton fluxes in mitochondria.
Collapse
Affiliation(s)
- Marcelo J Berardi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - James J Chou
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; National Center for Protein Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
237
|
Vera LM, Montoya A, Pujante IM, Pérez-Sánchez J, Calduch-Giner JA, Mancera JM, Moliner J, Sánchez-Vázquez FJ. Acute stress response in gilthead sea bream (Sparus aurata L.) is time-of-day dependent: Physiological and oxidative stress indicators. Chronobiol Int 2014; 31:1051-61. [PMID: 25102424 DOI: 10.3109/07420528.2014.945646] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since fish show daily rhythms in most physiological functions, it should not be surprising that stressors may have different effects depending on the timing of exposure. In this study, we investigated the influence of time of day on the stress responses, at both physiological and cellular levels, in gilthead sea bream (Sparus aurata L.) submitted to air exposure for 30 s and then returned to their tank. One hour after air exposure, blood, hypothalamus and liver samples were taken. Six fish per experimental group (control and stressed) were sampled every 4 h during a 24-h cycle. Fish were fed in the middle of the light cycle (ML) and locomotor activity rhythms were recorded using infrared photocells to determine their daily activity pattern of behaviour, which showed a peak around feeding time in all fish. In the control group, cortisol levels did not show daily rhythmicity, whereas in the stressed fish, a daily rhythm of plasma cortisol was observed, being the average values higher than in the control group, with increased differences during the dark phase. Blood glucose showed daily rhythmicity in the control group but not in the stressed one which also showed higher values at all sampling points. In the hypothalamus of control fish, a daily rhythm of corticotropin-releasing hormone (crh) gene expression was observed, with the acrophase at the beginning of the light phase. However, in the stressed fish, this rhythm was abolished. The expression of crh-binding protein (crhbp) showed a peak at the end of the dark phase in the control group, whereas in the stressed sea bream, this peak was found at ML. Regarding hepatic gene expression of oxidative stress biomarkers: (i) cytochrome c oxidase 4 showed daily rhythmicity in both control and stressed fish, with the acrophases located around ML, (ii) peroxiredoxin (prdx) 3 and 5 (prdx5) only presented daily rhythmicity of expression in the stressed fish, with the acrophase located at the beginning of the light cycle and (iii) uncoupling protein 1 showed significant differences between sampling points only in the control group, with significantly higher expression at the beginning of the dark phase. Taken together, these results indicate that stress response in gilthead sea bream is time-dependent as cortisol level rose higher at night, and that different rhythmic mechanisms interplay in the control of neuroendocrine and cellular stress responses.
Collapse
Affiliation(s)
- Luisa María Vera
- Department of Physiology, Faculty of Biology, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia , Murcia , Spain
| | | | | | | | | | | | | | | |
Collapse
|
238
|
Uncoupling protein 2 regulates palmitic acid-induced hepatoma cell autophagy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:810401. [PMID: 25512910 PMCID: PMC4143590 DOI: 10.1155/2014/810401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/29/2014] [Accepted: 06/30/2014] [Indexed: 01/11/2023]
Abstract
Mitochondrial uncoupling protein 2 (UCP2) is suggested to have a role in the development of nonalcoholic steatohepatitis (NASH). However, the mechanism remains unclear. Autophagy is an important mediator of many pathological responses. This study aims to investigate the relationship between UCP2 and hepatoma cells autophagy in palmitic acid- (PA-) induced lipotoxicity. H4IIE cells were treated with palmitic acid (PA), and cell autophagy and apoptosis were examined. UCP2 expression, in association with LC3-II and caspase-3, which are indicators of cell autophagy and apoptosis, respectively,was measured. Results demonstrated that UCP2 was associated with autophagy during PA-induced hepatic carcinoma cells injury. Tests on reactive oxygen species (ROS) showed that UCP2 overexpression strongly decreases PA-induced ROS production and apoptosis. Conversely, UCP2 inhibition by genipin or UCP2 mRNA silencing enhances PA-induced ROS production and apoptosis. Autophagy partially participates in this progress. Moreover, UCP2 was associated with ATP synthesis during PA-induced autophagy. In conclusion, increasing UCP2 expression in hepatoma cells may contribute to cell autophagy and antiapoptotic as result of fatty acid injury. Our results may bring new insights for potential NASH therapies.
Collapse
|
239
|
Emmerstorfer A, Wriessnegger T, Hirz M, Pichler H. Overexpression of membrane proteins from higher eukaryotes in yeasts. Appl Microbiol Biotechnol 2014; 98:7671-98. [PMID: 25070595 DOI: 10.1007/s00253-014-5948-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 02/08/2023]
Abstract
Heterologous expression and characterisation of the membrane proteins of higher eukaryotes is of paramount interest in fundamental and applied research. Due to the rather simple and well-established methods for their genetic modification and cultivation, yeast cells are attractive host systems for recombinant protein production. This review provides an overview on the remarkable progress, and discusses pitfalls, in applying various yeast host strains for high-level expression of eukaryotic membrane proteins. In contrast to the cell lines of higher eukaryotes, yeasts permit efficient library screening methods. Modified yeasts are used as high-throughput screening tools for heterologous membrane protein functions or as benchmark for analysing drug-target relationships, e.g., by using yeasts as sensors. Furthermore, yeasts are powerful hosts for revealing interactions stabilising and/or activating membrane proteins. We also discuss the stress responses of yeasts upon heterologous expression of membrane proteins. Through co-expression of chaperones and/or optimising yeast cultivation and expression strategies, yield-optimised hosts have been created for membrane protein crystallography or efficient whole-cell production of fine chemicals.
Collapse
Affiliation(s)
- Anita Emmerstorfer
- ACIB-Austrian Centre of Industrial Biotechnology, Petersgasse 14, 8010, Graz, Austria
| | | | | | | |
Collapse
|
240
|
Alves-Bezerra M, Cosentino-Gomes D, Vieira LP, Rocco-Machado N, Gondim KC, Meyer-Fernandes JR. Identification of uncoupling protein 4 from the blood-sucking insect Rhodnius prolixus and its possible role on protection against oxidative stress. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 50:24-33. [PMID: 24746771 DOI: 10.1016/j.ibmb.2014.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/14/2014] [Accepted: 03/24/2014] [Indexed: 06/03/2023]
Abstract
Uncoupling proteins (UCPs) play a critical role in the control of the mitochondrial membrane potential (ΔΨm) due to their ability to dissipate the proton gradient, which results in the uncoupling of mitochondrial respiration from ATP production. Most reactive oxygen species generation in mitochondria occurs in complex III, due to an increase of semiquinone (Q(-)) half-life. When active, UCPs can account as a potential antioxidant system by decreasing ΔΨm and increasing mitochondrial respiration, thus reducing Q(-) life time. The hematophagous insect Rhodnius prolixus, a vector of Chagas disease, is exposed to a huge increase in oxidative stress after a blood meal because of the hydrolysis of hemoglobin and the release of the cytotoxic heme molecule. Although some protective mechanisms were already described for this insect and other hematophagous arthropods, the putative role of UCP proteins as antioxidants in this context has not been explored. In this report, two genes encoding UCP proteins (RpUcp4 and RpUcp5) were identified in the R. prolixus genome. RpUcp4 is the predominant transcript in most analyzed organs, and both mRNA and protein expression are upregulated (13- and 3-fold increase, respectively) in enterocytes the first day after the blood feeding. The increase in UCP4 expression is coincident with the decrease in hydrogen peroxide (H2O2) generation by midgut cells. Furthermore, in mitochondria isolated from enterocytes, the modulation of UCP activity by palmitic acid and GDP resulted in altered ΔΨm, as well as modulation of H2O2 generation rates. These results indicate that R. prolixus UCP4 may function in an antioxidation mechanism to protect the midgut cells against oxidative damage caused by blood digestion.
Collapse
Affiliation(s)
- Michele Alves-Bezerra
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniela Cosentino-Gomes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lisvane P Vieira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathália Rocco-Machado
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Katia C Gondim
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Brazil
| | - José R Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Brazil.
| |
Collapse
|
241
|
Tang SE, Wu CP, Wu SY, Peng CK, Perng WC, Kang BH, Chu SJ, Huang KL. Stanniocalcin-1 ameliorates lipopolysaccharide-induced pulmonary oxidative stress, inflammation, and apoptosis in mice. Free Radic Biol Med 2014; 71:321-331. [PMID: 24685991 DOI: 10.1016/j.freeradbiomed.2014.03.034] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/20/2014] [Accepted: 03/22/2014] [Indexed: 12/20/2022]
Abstract
Stanniocalcin-1 (STC1) is an endogenous glycoprotein whose anti-inflammatory effects occur through induction of uncoupling proteins to reduce oxidative stress. In this study, we tested the hypothesis that exogenous recombinant human STC1 (rhSTC1) protects against lipopolysaccharide (LPS)-induced acute lung injury in mice. Anesthetized C57BL/6 mice underwent intratracheal spraying of LPS (20 µg/10 g body wt), and lung injury was assessed 24h later by analyzing pulmonary edema, bronchoalveolar lavage fluid, and lung histopathology. Lung inflammation, oxidative stress, and expression of STC1 and its downstream uncoupling protein 2 (UCP2) were analyzed at specific time points. Expression of UCP2 was suppressed initially but was subsequently upregulated after STC1 elevation in response to intratracheal administration of LPS. Intratracheal rhSTC1 treatment 1h before or after LPS spraying significantly attenuated pulmonary inflammation, oxidative stress, cell apoptosis, and acute lung injury. Pretreatment with STC1 short interfering RNA 48 h before LPS spraying inhibited the expression of STC1 and UCP2 and significantly increased the extent of lung injury. These findings suggest that STC1 is an endogenous stress protein that may counteract LPS-induced lung injury by inhibiting the inflammatory cascade and inducing antioxidant and antiapoptotic mechanisms. However, the potential clinical application of STC1 and the direct linkage between UCP2 and LPS-induced lung injury remain to be further investigated.
Collapse
Affiliation(s)
- Shih-En Tang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Landseed Hospital, Taoyuan, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei 114, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Wann-Cherng Perng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Bor-Hwang Kang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei 114, Taiwan
| | - Shi-Jye Chu
- Division of Rheumatology, Immunology, and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan.
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei 114, Taiwan.
| |
Collapse
|
242
|
Bermejo-Nogales A, Calduch-Giner JA, Pérez-Sánchez J. Tissue-specific gene expression and functional regulation of uncoupling protein 2 (UCP2) by hypoxia and nutrient availability in gilthead sea bream (Sparus aurata): implications on the physiological significance of UCP1-3 variants. FISH PHYSIOLOGY AND BIOCHEMISTRY 2014; 40:751-762. [PMID: 24154671 DOI: 10.1007/s10695-013-9882-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 10/12/2013] [Indexed: 06/02/2023]
Abstract
The aim of this study was to assess in an integrative manner the physiological regulation of uncoupling protein 2 (UCP2) in gilthead sea bream. A contig of 1,325 nucleotides in length with an open reading frame of 307 amino acids was recognized as UCP2 after searches in our transcriptome reference database ( http://www.nutrigroup-iats.org/seabreamdb ). Gene expression mapping by quantitative real-time PCR revealed a ubiquitous profile that clearly differs from that of UCP1 and UCP3 variants with the greatest abundance in liver and white skeletal muscle, respectively. The greatest abundance of UCP2 transcripts was found in the heart, with a relatively high expression level in blood cells, where UCP1 and UCP3 transcripts were practically undetectable. Functional studies revealed that UCP2 mRNA expression remains either unaltered or up-regulated upon feed restriction in glycolytic (white skeletal muscle) and highly oxidative muscle tissues (heart and red skeletal muscle), respectively. In contrast, exposure to hypoxic conditions (18-19% oxygen saturation) markedly down-regulated the UCP2 mRNA expression in blood cells in a cellular environment with increased haematocrit, blood haemoglobin content, and circulating levels of glucose and lactate, and total plasma antioxidant activity. These findings demonstrated that UCP2 expression is highly regulated at the transcriptional level, arising this UCP variant as an important piece of the complex trade-off between metabolic and redox sensors. This feature would avoid the activation of futile cycles of energy wastage if changes in tissue oxidative and antioxidant metabolic capabilities are able to maintain the production of reactive oxygen species at a low regulated level.
Collapse
Affiliation(s)
- Azucena Bermejo-Nogales
- Nutrigenomics and Fish Growth Endocrinology Group, Department of Biology, Culture and Pathology of Marine Species, Institute of Aquaculture Torre de la Sal, CSIC, 12595, Ribera de Cabanes, Castellón, Spain
| | | | | |
Collapse
|
243
|
Abstract
The field of mitochondrial ion channels has recently seen substantial progress, including the molecular identification of some of the channels. An integrative approach using genetics, electrophysiology, pharmacology, and cell biology to clarify the roles of these channels has thus become possible. It is by now clear that many of these channels are important for energy supply by the mitochondria and have a major impact on the fate of the entire cell as well. The purpose of this review is to provide an up-to-date overview of the electrophysiological properties, molecular identity, and pathophysiological functions of the mitochondrial ion channels studied so far and to highlight possible therapeutic perspectives based on current information.
Collapse
|
244
|
Barreto P, Okura VK, Neshich IAP, Maia IDG, Arruda P. Overexpression of UCP1 in tobacco induces mitochondrial biogenesis and amplifies a broad stress response. BMC PLANT BIOLOGY 2014; 14:144. [PMID: 24886177 PMCID: PMC4046140 DOI: 10.1186/1471-2229-14-144] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 05/23/2014] [Indexed: 05/19/2023]
Abstract
BACKGROUND Uncoupling protein one (UCP1) is a mitochondrial inner membrane protein capable of uncoupling the electrochemical gradient from adenosine-5'-triphosphate (ATP) synthesis, dissipating energy as heat. UCP1 plays a central role in nonshivering thermogenesis in the brown adipose tissue (BAT) of hibernating animals and small rodents. A UCP1 ortholog also occurs in plants, and aside from its role in uncoupling respiration from ATP synthesis, thereby wasting energy, it plays a beneficial role in the plant response to several abiotic stresses, possibly by decreasing the production of reactive oxygen species (ROS) and regulating cellular redox homeostasis. However, the molecular mechanisms by which UCP1 is associated with stress tolerance remain unknown. RESULTS Here, we report that the overexpression of UCP1 increases mitochondrial biogenesis, increases the uncoupled respiration of isolated mitochondria, and decreases cellular ATP concentration. We observed that the overexpression of UCP1 alters mitochondrial bioenergetics and modulates mitochondrial-nuclear communication, inducing the upregulation of hundreds of nuclear- and mitochondrial-encoded mitochondrial proteins. Electron microscopy analysis showed that these metabolic changes were associated with alterations in mitochondrial number, area and morphology. Surprisingly, UCP1 overexpression also induces the upregulation of hundreds of stress-responsive genes, including some involved in the antioxidant defense system, such as superoxide dismutase (SOD), glutathione peroxidase (GPX) and glutathione-S-transferase (GST). As a consequence of the increased UCP1 activity and increased expression of oxidative stress-responsive genes, the UCP1-overexpressing plants showed reduced ROS accumulation. These beneficial metabolic effects may be responsible for the better performance of UCP1-overexpressing lines in low pH, high salt, high osmolarity, low temperature, and oxidative stress conditions. CONCLUSIONS Overexpression of UCP1 in the mitochondrial inner membrane induced increased uncoupling respiration, decreased ROS accumulation under abiotic stresses, and diminished cellular ATP content. These events may have triggered the expression of mitochondrial and stress-responsive genes in a coordinated manner. Because these metabolic alterations did not impair plant growth and development, UCP1 overexpression can potentially be used to create crops better adapted to abiotic stress conditions.
Collapse
Affiliation(s)
- Pedro Barreto
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas (UNICAMP), 13083-875 Campinas, SP, Brazil
| | - Vagner Katsumi Okura
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas (UNICAMP), 13083-875 Campinas, SP, Brazil
| | - Izabella Agostinho Pena Neshich
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas (UNICAMP), 13083-875 Campinas, SP, Brazil
| | - Ivan de Godoy Maia
- Departamento de Genética, Instituto de Biociências, UNESP, 18618-970 Botucatu, SP, Brazil
| | - Paulo Arruda
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas (UNICAMP), 13083-875 Campinas, SP, Brazil
- Departamento de Genética e Evolução, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), 13083-875 Campinas, SP, Brazil
| |
Collapse
|
245
|
Donadelli M, Dando I, Fiorini C, Palmieri M. UCP2, a mitochondrial protein regulated at multiple levels. Cell Mol Life Sci 2014; 71:1171-90. [PMID: 23807210 PMCID: PMC11114077 DOI: 10.1007/s00018-013-1407-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/16/2013] [Accepted: 06/10/2013] [Indexed: 12/11/2022]
Abstract
An ever-increasing number of studies highlight the role of uncoupling protein 2 (UCP2) in a broad range of physiological and pathological processes. The knowledge of the molecular mechanisms of UCP2 regulation is becoming fundamental in both the comprehension of UCP2-related physiological events and the identification of novel therapeutic strategies based on UCP2 modulation. The study of UCP2 regulation is a fast-moving field. Recently, several research groups have made a great effort to thoroughly understand the various molecular mechanisms at the basis of UCP2 regulation. In this review, we describe novel findings concerning events that can occur in a concerted manner at various levels: Ucp2 gene mutation (single nucleotide polymorphisms), UCP2 mRNA and protein expression (transcriptional, translational, and protein turn-over regulation), UCP2 proton conductance (ligands and post-transcriptional modifications), and nutritional and pharmacological regulation of UCP2.
Collapse
Affiliation(s)
- Massimo Donadelli
- Section of Biochemistry, Deparment of Life and Reproduction Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy,
| | | | | | | |
Collapse
|
246
|
JEŽEK P, OLEJÁR T, SMOLKOVÁ K, JEŽEK J, DLASKOVÁ A, PLECITÁ-HLAVATÁ L, ZELENKA J, ŠPAČEK T, ENGSTOVÁ H, PAJUELO REGUERA D, JABŮREK M. Antioxidant and Regulatory Role of Mitochondrial Uncoupling Protein UCP2 in Pancreatic β-cells. Physiol Res 2014; 63:S73-91. [DOI: 10.33549/physiolres.932633] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Research on brown adipose tissue and its hallmark protein, mitochondrial uncoupling protein UCP1, has been conducted for half a century and has been traditionally studied in the Institute of Physiology (AS CR, Prague), likewise UCP2 residing in multiple tissues for the last two decades. Our group has significantly contributed to the elucidation of UCP uncoupling mechanism, fully dependent on free fatty acids (FFAs) within the inner mitochondrial membrane. Now we review UCP2 physiological roles emphasizing its roles in pancreatic β-cells, such as antioxidant role, possible tuning of redox homeostasis (consequently UCP2 participation in redox regulations), and fine regulation of glucose-stimulated insulin secretion (GSIS). For example, NADPH has been firmly established as being a modulator of GSIS and since UCP2 may influence redox homeostasis, it likely affects NADPH levels. We also point out the role of phospholipase iPLA2 isoform in providing FFAs for the UCP2 antioxidant function. Such initiation of mild uncoupling hypothetically precedes lipotoxicity in pancreatic β-cells until it reaches the pathological threshold, after which the antioxidant role of UCP2 can be no more cell-protective, for example due to oxidative stress-accumulated mutations in mtDNA. These mechanisms, together with impaired autocrine insulin function belong to important causes of Type 2 diabetes etiology.
Collapse
Affiliation(s)
- P. JEŽEK
- Department of Membrane Transport Biophysics, Institute of Physiology Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Uncoupling protein 2 and 4 expression pattern during stem cell differentiation provides new insight into their putative function. PLoS One 2014; 9:e88474. [PMID: 24523901 PMCID: PMC3921169 DOI: 10.1371/journal.pone.0088474] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/08/2014] [Indexed: 11/19/2022] Open
Abstract
Apart from the first family member, uncoupling protein 1 (UCP1), the functions of other UCPs (UCP2-UCP5) are still unknown. In analyzing our own results and those previously published by others, we have assumed that UCP's cellular expression pattern coincides with a specific cell metabolism and changes if the latter is altered. To verify this hypothesis, we analyzed the expression of UCP1-5 in mouse embryonic stem cells before and after their differentiation to neurons. We have shown that only UCP2 is present in undifferentiated stem cells and it disappears simultaneously with the initiation of neuronal differentiation. In contrast, UCP4 is simultaneously up-regulated together with typical neuronal marker proteins TUJ-1 and NeuN during mESC differentiation in vitro as well as during murine brain development in vivo. Notably, several tested cell lines express UCP2, but not UCP4. In line with this finding, neuroblastoma cells that display metabolic features of tumor cells express UCP2, but not UCP4. UCP2's occurrence in cancer, immunological and stem cells indicates that UCP2 is present in cells with highly proliferative potential, which have a glycolytic type of metabolism as a common feature, whereas UCP4 is strongly associated with non-proliferative highly differentiated neuronal cells.
Collapse
|
248
|
Uncoupling protein 2 deficiency aggravates astrocytic endoplasmic reticulum stress and nod-like receptor protein 3 inflammasome activation. Neurobiol Aging 2014; 35:421-30. [DOI: 10.1016/j.neurobiolaging.2013.08.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 08/02/2013] [Accepted: 08/16/2013] [Indexed: 01/07/2023]
|
249
|
Park EY, Kim MH, Kim EH, Lee EK, Park IS, Yang DC, Jun HS. Efficacy Comparison of Korean Ginseng and American Ginseng on Body Temperature and Metabolic Parameters. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:173-87. [DOI: 10.1142/s0192415x14500128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ginseng has beneficial effects in cancer, diabetes and aging. There are two main varieties of ginseng: Panax ginseng (Korean ginseng) and Panax quinquefolius (American ginseng). There are anecdotal reports that American ginseng helps reduce body temperature, whereas Korean ginseng improves blood circulation and increases body temperature; however, their respective effects on body temperature and metabolic parameters have not been studied. We investigated body temperature and metabolic parameters in mice using a metabolic cage. After administering ginseng extracts acutely (single dose of 1000 mg/kg) or chronically (200 mg/kg/day for four weeks), core body temperature, food intake, oxygen consumption and activity were measured, as well as serum levels of pyrogen-related factors and mRNA expression of metabolic genes. Acute treatment with American ginseng reduced body temperature compared with PBS-treated mice during the night; however, there was no significant effect of ginseng treatment on body temperature after four weeks of treatment. VO 2, VCO 2, food intake, activity and energy expenditure were unchanged after both acute and chronic ginseng treatment compared with PBS treatment. In acutely treated mice, serum thyroxin levels were reduced by red and American ginseng, and the serum prostaglandin E2 level was reduced by American ginseng. In chronically treated mice, red and white ginseng reduced thyroxin levels. We conclude that Korean ginseng does not stimulate metabolism in mice, whereas a high dose of American ginseng may reduce night-time body temperature and pyrogen-related factors.
Collapse
Affiliation(s)
- Eun-Young Park
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Mi-Hwi Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Eung-Hwi Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Eun-Kyu Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - In-Sun Park
- Department of Pathology, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Duck-Choon Yang
- Korea Ginseng Center for Most Valuable Products and Ginseng Genetic Resource Bank, Kyung Hee University, Korea
| | - Hee-Sook Jun
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Gachon Medical Research Institute, Gil Hospital, Incheon, Korea
| |
Collapse
|
250
|
Hashemi M, Rezaei H, Kaykhaei MA, Taheri M. A 45-bp insertion/deletion polymorphism of UCP2 gene is associated with metabolic syndrome. J Diabetes Metab Disord 2014; 13:12. [PMID: 24398006 PMCID: PMC3937167 DOI: 10.1186/2251-6581-13-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 12/21/2013] [Indexed: 12/21/2022]
Abstract
Background Metabolic syndrome (MeS) is being recognized as a risk factor for insulin resistance and cardiovascular disease. The present study was aimed to find out the possible association between 45-bp I/D polymorphism of uncoupling protein 2 (UCP2) and MeS. Methods DNA was extracted from peripheral blood of 151 subjects with and 149 subjects without MeS. 45-bp I/D variant of UCP2 was detected using polymerase chain reaction (PCR). Results Our finding showed that 45-bp I/D polymorphism was associated with protection against MeS (OR = 0.56, 95% CI = 0.34-0.92, p = 0.020 D/I vs DD and OR = 0.54, 95% CI = 0.34-0.86, p = 0.009; D/I + I/I vs D/D). The I allele decreased the risk of MeS (OR = 0.62, 95% CI = 0.44-0.90, p = 0.011) in comparison with D allele. Conclusion In conclusion, our result suggests that 45-bp I/D polymorphism is associated with the risk of MeS, which remains to be cleared.
Collapse
Affiliation(s)
- Mohammad Hashemi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | | | | | | |
Collapse
|