151
|
Tenta R, Fragopoulou E, Tsoukala M, Xanthopoulou M, Skyrianou M, Pratsinis H, Kletsas D. Antiproliferative Effects of Red and White Wine Extracts in PC-3 Prostate Cancer Cells. Nutr Cancer 2017; 69:952-961. [DOI: 10.1080/01635581.2017.1340489] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Roxane Tenta
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | | | - Magafoula Tsoukala
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | | | - Maria Skyrianou
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Harris Pratsinis
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| |
Collapse
|
152
|
Bharath LP, Cho JM, Park SK, Ruan T, Li Y, Mueller R, Bean T, Reese V, Richardson RS, Cai J, Sargsyan A, Pires K, Anandh Babu PV, Boudina S, Graham TE, Symons JD. Endothelial Cell Autophagy Maintains Shear Stress-Induced Nitric Oxide Generation via Glycolysis-Dependent Purinergic Signaling to Endothelial Nitric Oxide Synthase. Arterioscler Thromb Vasc Biol 2017; 37:1646-1656. [PMID: 28684613 DOI: 10.1161/atvbaha.117.309510] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/19/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Impaired endothelial cell (EC) autophagy compromises shear stress-induced nitric oxide (NO) generation. We determined the responsible mechanism. APPROACH AND RESULTS On autophagy compromise in bovine aortic ECs exposed to shear stress, a decrease in glucose uptake and EC glycolysis attenuated ATP production. We hypothesized that decreased glycolysis-dependent purinergic signaling via P2Y1 (P2Y purinoceptor 1) receptors, secondary to impaired autophagy in ECs, prevents shear-induced phosphorylation of eNOS (endothelial nitric oxide synthase) at its positive regulatory site S1117 (p-eNOSS1177) and NO generation. Maneuvers that restore glucose transport and glycolysis (eg, overexpression of GLUT1 [glucose transporter 1]) or purinergic signaling (eg, addition of exogenous ADP) rescue shear-induced p-eNOSS1177 and NO production in ECs with impaired autophagy. Conversely, inhibiting glucose transport via GLUT1 small interfering RNA, blocking purinergic signaling via ectonucleotidase-mediated ATP/ADP degradation (eg, apyrase), or inhibiting P2Y1 receptors using pharmacological (eg, MRS2179 [2'-deoxy-N6-methyladenosine 3',5'-bisphosphate tetrasodium salt]) or genetic (eg, P2Y1-receptor small interfering RNA) procedures inhibit shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Supporting a central role for PKCδT505 (protein kinase C delta T505) in relaying the autophagy-dependent purinergic-mediated signal to eNOS, we find that (1) shear stress-induced activating phosphorylation of PKCδT505 is negated by inhibiting autophagy, (2) shear-induced p-eNOSS1177 and NO generation are restored in autophagy-impaired ECs via pharmacological (eg, bryostatin) or genetic (eg, constitutively active PKCδ) activation of PKCδT505, and (3) pharmacological (eg, rottlerin) and genetic (eg, PKCδ small interfering RNA) PKCδ inhibition prevents shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Key nodes of dysregulation in this pathway on autophagy compromise were revealed in human arterial ECs. CONCLUSIONS Targeted reactivation of purinergic signaling and PKCδ has strategic potential to restore compromised NO generation in pathologies associated with suppressed EC autophagy.
Collapse
Affiliation(s)
- Leena P Bharath
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jae Min Cho
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Seul-Ki Park
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ting Ruan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Youyou Li
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Robert Mueller
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Tyler Bean
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Van Reese
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Russel S Richardson
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jinjin Cai
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ashot Sargsyan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Karla Pires
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Pon Velayutham Anandh Babu
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Sihem Boudina
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Timothy E Graham
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - J David Symons
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.).
| |
Collapse
|
153
|
Macias-Ceja DC, Cosín-Roger J, Ortiz-Masiá D, Salvador P, Hernández C, Esplugues JV, Calatayud S, Barrachina MD. Stimulation of autophagy prevents intestinal mucosal inflammation and ameliorates murine colitis. Br J Pharmacol 2017; 174:2501-2511. [PMID: 28500644 DOI: 10.1111/bph.13860] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Defective autophagy contributes to the pathogenesis of inflammatory disorders such as inflammatory bowel disease and there are interactions between autophagy and inflammation. Here we have analysed the effects of autophagy stimulators on murine colitis. EXPERIMENTAL APPROACH Mice were treated with intrarectal administration of 2,4,6-trinitrobenzenesulfonic acid (TNBS) (3.5 mg·20 g-1 ) and body weight was measured daily. Histological damage was scored 2 or 4 days after treatment. Some mice received trehalose (3% in drinking water 3 weeks before TNBS administration) or a daily administration of rapamycin (1.25 mg·kg-1 , i.p.), betanin (1 g·kg-1 , i.p.) or betanin + 3-methyladenine (3MA) (10 mg·kg-1 , i.p.). Protein levels of p-mTOR, p62, LC3, BCL10, NFκB, IκBα and p-IκBα in mucosa were determined by Western blots and mRNA expression of TNFα, IL1β, IL6, IL10, COX2, CCR7, CD11c, inducible NOS and CD86 by qRT-PCR. KEY RESULTS Impaired autophagy associated with body weight loss and intestinal damage was detected in the mucosa of TNBS-treated mice. Administration of trehalose, rapamycin or betanin prevented the impaired autophagic flux induced by TNBS and decreased mucosal protein levels of BCL10, p-IκBα and NFκB-p65 and the expression of pro-inflammatory cytokines and M1 macrophage markers. Blockade of autophagosome formation by treatment with 3MA, prevented the reduction in protein levels of p62, BCL10, p-IκBα and NFκB-p65 induced by betanin in TNBS-treated mice and weakened the protective effects of betanin on murine colitis. CONCLUSIONS AND IMPLICATIONS Pharmacological stimulation of mucosal autophagy reduced intestinal inflammation and improved murine colitis.
Collapse
Affiliation(s)
| | - Jesús Cosín-Roger
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Dolores Ortiz-Masiá
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain.,Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Pedro Salvador
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Carlos Hernández
- FISABIO, Hospital Dr. Peset, Valencia, Spain.,Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Juan V Esplugues
- FISABIO, Hospital Dr. Peset, Valencia, Spain.,Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Sara Calatayud
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - María D Barrachina
- Departamento de Farmacología and CIBERehd, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
154
|
Li MX, Mu DZ. [Mitophagy and nervous system disease]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:724-729. [PMID: 28606244 PMCID: PMC7390300 DOI: 10.7499/j.issn.1008-8830.2017.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/15/2017] [Indexed: 06/07/2023]
Abstract
Mitophagy is a process during which the cell selectively removes the mitochondria via the mechanism of autophagy. It is crucial to the functional completeness of the whole mitochondrial network and determines cell survival and death. On the one hand, the damaged mitochondria releases pro-apoptotic factors which induce cell apoptosis; on the other hand, the damaged mitochondria eliminates itself via autophagy, which helps to maintain cell viability. Mitophagy is of vital importance for the development and function of the nervous system. Neural cells rely on autophagy to control protein quality and eliminate the damaged mitochondria, and under normal circumstances, mitophagy can protect the neural cells. Mutations in genes related to mitophagy may cause the development and progression of neurodegenerative diseases. An understanding of the role of mitophagy in nervous system diseases may provide new theoretical bases for clinical treatment. This article reviews the research advances in the relationship between mitophagy and different types of nervous system diseases.
Collapse
Affiliation(s)
- Ming-Xi Li
- Department of Pediatrics, West China Second Hospital, Sichuan University/Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education/Key Laboratory of Development and Related Diseases of Women and Children, Chengdu 610041, China.
| | | |
Collapse
|
155
|
Forte M, Palmerio S, Yee D, Frati G, Sciarretta S. Functional Role of Nox4 in Autophagy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:307-326. [DOI: 10.1007/978-3-319-55330-6_16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
156
|
Reid ES, Williams H, Anderson G, Benatti M, Chong K, James C, Ocaka L, Hemingway C, Little D, Brown R, Parker A, Holden S, Footitt E, Rahman S, Gissen P, Mills PB, Clayton PT. Mutations in SLC25A22: hyperprolinaemia, vacuolated fibroblasts and presentation with developmental delay. J Inherit Metab Dis 2017; 40:385-394. [PMID: 28255779 PMCID: PMC5393281 DOI: 10.1007/s10545-017-0025-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/07/2017] [Accepted: 02/03/2017] [Indexed: 12/19/2022]
Abstract
Mutations in SLC25A22 are known to cause neonatal epileptic encephalopathy and migrating partial seizures in infancy. Using whole exome sequencing we identified four novel SLC25A22 mutations in six children from three families. Five patients presented clinical features similar to those in the literature including hypotonia, refractory neonatal-onset seizures and developmental delay. However, the sixth patients presented atypically with isolated developmental delay, developing late-onset (absence) seizures only at 7 years of age. Abnormal metabolite levels have not been documented in the nine patients described previously. One patient in our series was referred to the metabolic clinic because of persistent hyperprolinaemia and another three had raised plasma proline when tested. Analysis of the post-prandial plasma amino acid response in one patient showed abnormally high concentrations of several amino acids. This suggested that, in the fed state, when amino acids are the preferred fuel for the liver, trans-deamination of amino acids requires transportation of glutamate into liver mitochondria by SLC25A22 for deamination by glutamate dehydrogenase; SLC25A22 is an important mitochondrial glutamate transporter in liver as well as in brain. Electron microscopy of patient fibroblasts demonstrated widespread vacuolation containing neutral and phospho-lipids as demonstrated by Oil Red O and Sudan Black tinctorial staining; this might be explained by impaired activity of the proline/pyrroline-5-carboxylate (P5C) shuttle if SLC25A22 transports pyrroline-5-carboxylate/glutamate-γ-semialdehyde as well as glutamate.
Collapse
Affiliation(s)
- Emma S Reid
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Hywel Williams
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Glenn Anderson
- Histopathology Department, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Malika Benatti
- Histopathology Department, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Kling Chong
- Radiology Department, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Chela James
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Louise Ocaka
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Cheryl Hemingway
- Neurology Department, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Daniel Little
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
- The Medical Research Council Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Richard Brown
- Peterborough and Stamford Hospitals NHS Foundation Trust, Edith Cavell Campus, Bretton Gate, Peterborough, PE3 9GZ, UK
| | - Alasdair Parker
- Child Development Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Simon Holden
- Clinical Genetics, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Emma Footitt
- Metabolic Medicine Department, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Shamima Rahman
- Metabolic Medicine Department, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
- Mitochondrial Research Group, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Paul Gissen
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
- The Medical Research Council Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
- Metabolic Medicine Department, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Philippa B Mills
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Peter T Clayton
- Centre for Translational Omics, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
157
|
Wu G, Liu XX, Lu NN, Liu QB, Tian Y, Ye WF, Jiang GJ, Tao RR, Han F, Lu YM. Endothelial ErbB4 deficit induces alterations in exploratory behavior and brain energy metabolism in mice. CNS Neurosci Ther 2017; 23:510-517. [PMID: 28421673 DOI: 10.1111/cns.12695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/12/2017] [Accepted: 03/15/2017] [Indexed: 12/17/2022] Open
Abstract
AIMS The receptor tyrosine kinase ErbB4 is present throughout the primate brain and has a distinct functional profile. In this study, we investigate the potential role of endothelial ErbB4 receptor signaling in the brain. RESULTS Here, we show that the endothelial cell-specific deletion of ErbB4 induces decreased exploratory behavior in adult mice. However, the water maze task for spatial memory and the memory reconsolidation test reveal no changes; additionally, we observe no impairment in CaMKII phosphorylation in Cdh5Cre;ErbB4f/f mice, which indicates that the endothelial ErbB4 deficit leads to decreased exploratory activity rather than direct memory deficits. Furthermore, decreased brain metabolism, which was measured using micro-positron emission tomography, is observed in the Cdh5Cre;ErbB4f/f mice. Consistently, the immunoblot data demonstrate the downregulation of brain Glut1, phospho-ULK1 (Ser555), and TIGAR in the endothelial ErbB4 conditional knockout mice. Collectively, our findings suggest that endothelial ErbB4 plays a critical role in regulating brain function, at least in part, through maintaining normal brain energy homeostasis. CONCLUSIONS Targeting ErbB4 or the modulation of endothelial ErbB4 signaling may represent a rational pharmacological approach to treat neurological disorders.
Collapse
Affiliation(s)
- Gang Wu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Xiu-Xiu Liu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Nan-Nan Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qi-Bing Liu
- School of Pharmacy, Hainan Medical College, Haikou, China
| | - Yun Tian
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wei-Feng Ye
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Guo-Jun Jiang
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang, China
| | - Rong-Rong Tao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feng Han
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ying-Mei Lu
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| |
Collapse
|
158
|
Song X, Narzt MS, Nagelreiter IM, Hohensinner P, Terlecki-Zaniewicz L, Tschachler E, Grillari J, Gruber F. Autophagy deficient keratinocytes display increased DNA damage, senescence and aberrant lipid composition after oxidative stress in vitro and in vivo. Redox Biol 2017; 11:219-230. [PMID: 28012437 PMCID: PMC5192251 DOI: 10.1016/j.redox.2016.12.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 01/05/2023] Open
Abstract
Autophagy allows cells fundamental adaptations to metabolic needs and to stress. Using autophagic bulk degradation cells can clear crosslinked macromolecules and damaged organelles that arise under redox stress. Accumulation of such debris results in cellular dysfunction and is observed in aged tissue and senescent cells. Conversely, promising anti-aging strategies aim at inhibiting the mTOR pathway and thereby activating autophagy, to counteract aging associated damage. We have inactivated autophagy related 7 (Atg7), an essential autophagy gene, in murine keratinocytes (KC) and have found in an earlier study that this resulted in increased baseline oxidative stress and reduced capacity to degrade crosslinked proteins after oxidative ultraviolet stress. To investigate whether autophagy deficiency would promote cellular aging, we studied how Atg7 deficient (KO) and Atg7 bearing cells (WT) would respond to stress induced by paraquat (PQ), an oxidant drug commonly used to induce cellular senescence. Atg7 deficient KC displayed increased prostanoid signaling and a pro- mitotic gene expression signature as compared to the WT. After exposure to PQ, both WT and KO cells showed an inflammatory and stress-related transcriptomic response. However, the Atg7 deficient cells additionally showed drastic DNA damage- and cell cycle arrest signaling. Indeed, DNA fragmentation and -oxidation were strongly increased in the stressed Atg7 deficient cells upon PQ stress but also after oxidizing ultraviolet A irradiation. Damage associated phosphorylated histone H2AX (γH2AX) foci were increased in the nuclei, whereas expression of the nuclear lamina protein lamin B1 was strongly decreased. Similarly, in both, PQ treated mouse tail skin explants and in UVA irradiated mouse tail skin, we found a strong increase in γH2AX positive nuclei within the basal layer of Atg7 deficient epidermis. Atg7 deficiency significantly affected expression of lipid metabolic genes. Therefore we performed lipid profiling of keratinocytes which demonstrated a major dysregulation of cellular lipid metabolism. We found accumulation of autophagy agonisitic free fatty acids, whereas triglyceride levels were strongly decreased. Together, our data show that in absence of Atg7/autophagy the resistance of keratinocytes to intrinsic and environmental oxidative stress was severely impaired and resulted in DNA damage, cell cycle arrest and a disturbed lipid phenotype, all typical for premature cell aging.
Collapse
Affiliation(s)
- Xiuzu Song
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria; Department of Dermatology, The Third Hospital of Hangzhou, 38 Xihu Road, Hangzhou, Zhejiang, 310009, PR China
| | - Marie Sophie Narzt
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria
| | - Ionela Mariana Nagelreiter
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria
| | - Philipp Hohensinner
- Department of Internal Medicine II - Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Austria
| | - Erwin Tschachler
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria
| | - Johannes Grillari
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Muthgasse 18, 1190 Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Austria
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Austria.
| |
Collapse
|
159
|
Abstract
One of the fundamental traits of immune cells in rheumatoid arthritis (RA) is their ability to proliferate, a property shared with the joint-resident cells that form the synovial pannus. The building of biomass imposes high demands for energy and biosynthetic precursors, implicating metabolic control as a basic disease mechanism. During preclinical RA, when autoreactive T cells expand and immunological tolerance is broken, the main sites of disease are the secondary lymphoid tissues. Naive CD4+ T cells from patients with RA have a distinct metabolic signature, characterized by dampened glycolysis, low ATP levels and enhanced shunting of glucose into the pentose phosphate pathway. Equipped with high levels of NADPH and depleted of intracellular reactive oxygen species, such T cells hyperproliferate and acquire proinflammatory effector functions. During clinical RA, immune cells coexist with stromal cells in the acidic milieu of the inflamed joint. This microenvironment is rich in metabolic intermediates that are released into the extracellular space to shape cell-cell communication and the functional activity of tissue-resident cells. Increasing awareness of how metabolites regulate signalling pathways, guide post-translational modifications and condition the tissue microenvironment will help to connect environmental factors with the pathogenic behaviour of T cells in RA.
Collapse
|
160
|
Zheng S, Han F, Shi Y, Wen L, Han D. Single-Prolonged-Stress-Induced Changes in Autophagy-Related Proteins Beclin-1, LC3, and p62 in the Medial Prefrontal Cortex of Rats with Post-traumatic Stress Disorder. J Mol Neurosci 2017; 62:43-54. [PMID: 28341893 DOI: 10.1007/s12031-017-0909-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/08/2017] [Indexed: 12/22/2022]
Abstract
Autophagy, or type II programmed cell death, plays a crucial role in many nervous system diseases. However, few studies have examined the role of autophagy in post-traumatic stress disorder (PTSD), and the mechanisms underlying PTSD are poorly understood. The objective of this research was to explore the expression of three important autophagy-related proteins, Beclin-1, microtubule-associated protein 1 light chain 3 (LC3), and p62/SQSTM1 (p62), in the medial prefrontal cortex (mPFC) of an animal model of PTSD to identify changes in autophagic activity during PTSD pathogenesis. PTSD was induced in rats by exposure to a single-prolonged stress (SPS). The Morris water maze was used to assess cognitive changes in rats from the SPS and control groups. Transmission electron microscopy (TEM) was employed to observe mPFC morphological changes. Immunohistochemistry, immunofluorescence, and Western blotting techniques were used to detect expression of Beclin-1, LC3, and p62 in the mPFC. The Morris water maze test results showed that the escape latency time was increased and that the percent time in the target quadrant was decreased in the SPS group compared with that in the control group. Numerous visible autolysosomes in mPFC neurons were observed using TEM after SPS stimulation. Compared with that in the control group, the expression of Beclin-1 and the LC3-II/I ratio significantly decreased at 1 day, then increased and peaked at 7 days, and slightly decreased at 14 days after SPS stimulation, whereas the converse was found for p62 expression. In conclusion, dysregulation of autophagic activity in the mPFC may play a crucial role in PTSD pathogenesis.
Collapse
Affiliation(s)
- Shilei Zheng
- PTSD Laboratory, Department of Histology and Embryology, Institute of Pathology and Pathophysiology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.,Department of Radiology, First Affiliated Hospital of Jinzhou Medical University, No. 2 fifth Duan, Renmin Street, Jinzhou, Liaoning, 121001, China
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Institute of Pathology and Pathophysiology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Institute of Pathology and Pathophysiology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| | - Lili Wen
- PTSD Laboratory, Department of Histology and Embryology, Institute of Pathology and Pathophysiology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Dan Han
- PTSD Laboratory, Department of Histology and Embryology, Institute of Pathology and Pathophysiology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| |
Collapse
|
161
|
Marín de Mas I, Marín S, Pachón G, Rodríguez-Prados JC, Vizán P, Centelles JJ, Tauler R, Azqueta A, Selivanov V, López de Ceraín A, Cascante M. Unveiling the Metabolic Changes on Muscle Cell Metabolism Underlying p-Phenylenediamine Toxicity. Front Mol Biosci 2017; 4:8. [PMID: 28321398 PMCID: PMC5338303 DOI: 10.3389/fmolb.2017.00008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/09/2017] [Indexed: 12/15/2022] Open
Abstract
Rhabdomyolysis is a disorder characterized by acute damage of the sarcolemma of the skeletal muscle leading to release of potentially toxic muscle cell components into the circulation, most notably creatine phosphokinase (CK) and myoglobulin, and is frequently accompanied by myoglobinuria. In the present work, we evaluated the toxicity of p-phenylenediamine (PPD), a main component of hair dyes which is reported to induce rhabdomyolysis. We studied the metabolic effect of this compound in vivo with Wistar rats and in vitro with C2C12 muscle cells. To this aim we have combined multi-omic experimental measurements with computational approaches using model-driven methods. The integrative study presented here has unveiled the metabolic disorders associated to PPD exposure that may underlay the aberrant metabolism observed in rhabdomyolys disease. Animals treated with lower doses of PPD (10 and 20 mg/kg) showed depressed activity and myoglobinuria after 10 h of treatment. We measured the serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and creatine kinase (CK) in rats after 24, 48, and 72 h of PPD exposure. At all times, treatment with PPD at higher doses (40 and 60 mg/kg) showed an increase of AST and ALT, and also an increase of lactate dehydrogenase (LDH) and CK after 24 h. Blood packed cell volume and hemoglobin levels, as well as organs weight at 48 and 72 h, were also measured. No significant differences were observed in these parameters under any condition. PPD induce cell cycle arrest in S phase and apoptosis (40% or early apoptotic cells) on mus musculus mouse C2C12 cells after 24 h of treatment. Incubation of mus musculus mouse C2C12 cells with [1,2-13C2]-glucose during 24 h, subsequent quantification of 13C isotopologues distribution in key metabolites of glucose metabolic network and a computational fluxomic analysis using in-house developed software (Isodyn) showed that PPD is inhibiting glycolysis, non-oxidative pentose phosphate pathway, glycogen turnover, and ATPAse reaction leading to a reduction in ATP synthesis. These findings unveil the glucose metabolism collapse, which is consistent with a decrease in cell viability observed in PPD-treated C2C12 cells and with the myoglubinuria and other effects observed in Wistar Rats treated with PPD. These findings shed new light on muscle dysfunction associated to PPD exposure, opening new avenues for cost-effective therapies in Rhabdomyolysis disease.
Collapse
Affiliation(s)
- Igor Marín de Mas
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de BarcelonaBarcelona, Spain; Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research, Consejo Superior de Investigaciones CientíficasBarcelona, Spain
| | - Silvia Marín
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona Barcelona, Spain
| | - Gisela Pachón
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona Barcelona, Spain
| | - Juan C Rodríguez-Prados
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona Barcelona, Spain
| | - Pedro Vizán
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona Barcelona, Spain
| | - Josep J Centelles
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona Barcelona, Spain
| | - Romà Tauler
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research, Consejo Superior de Investigaciones Científicas Barcelona, Spain
| | - Amaya Azqueta
- Departamento de Farmacología y Toxicología, Facultad de Farmacia y Nutrición, Universidad de Navarra Pamplona, Spain
| | - Vitaly Selivanov
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona Barcelona, Spain
| | - Adela López de Ceraín
- Departamento de Farmacología y Toxicología, Facultad de Farmacia y Nutrición, Universidad de Navarra Pamplona, Spain
| | - Marta Cascante
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona Barcelona, Spain
| |
Collapse
|
162
|
Harnett MM, Pineda MA, Latré de Laté P, Eason RJ, Besteiro S, Harnett W, Langsley G. From Christian de Duve to Yoshinori Ohsumi: More to autophagy than just dining at home. Biomed J 2017; 40:9-22. [PMID: 28411887 PMCID: PMC6138802 DOI: 10.1016/j.bj.2016.12.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 12/26/2016] [Accepted: 12/28/2016] [Indexed: 12/21/2022] Open
Abstract
Christian de Duve first coined the expression “autophagy” during his seminal work on the discovery of lysosomes, which led to him being awarded the Nobel Prize in Physiology or Medicine in 1974. The term was adopted to distinguish degradation of intracellular components from the uptake and degradation of extracellular substances that he called “heterophagy”. Studies until the 1990s were largely observational/morphological-based until in 1993 Yoshinori Oshumi described a genetic screen in yeast undergoing nitrogen deprivation that led to the isolation of autophagy-defective mutants now better known as ATG (AuTophaGy-related) genes. The screen identified mutants that fell into 15 complementation groups implying that at least 15 genes were involved in the regulation of autophagy in yeast undergoing nutrient deprivation, but today, 41 yeast ATG genes have been described and many (though not all) have orthologues in humans. Attempts to identify the genetic basis of autophagy led to an explosion in its research and it's not surprising that in 2016 Yoshinori Oshumi was awarded the Nobel Prize in Physiology or Medicine. Our aim here is not to exhaustively review the ever-expanding autophagy literature (>60 papers per week), but to celebrate Yoshinori Oshumi's Nobel Prize by highlighting just a few aspects that are not normally extensively covered. In an accompanying mini-review we address the role of autophagy in early-diverging eukaryote parasites that like yeast, lack lysosomes and so use a digestive vacuole to degrade autophagosome cargo and also discuss how parasitized host cells react to infection by subverting regulation of autophagy.
Collapse
Affiliation(s)
- Margaret M Harnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, UK.
| | - Miguel A Pineda
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, UK
| | - Perle Latré de Laté
- Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France; The laboratory of Comparative Cell Biology of Apicomplexa, Medical Faculty of Paris-Descartes University, Sorbonne Paris City, France
| | - Russell J Eason
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, UK
| | - Sébastien Besteiro
- DIMNP, UMR CNRS 5235, Montpellier University, Place Eugène Bataillon, Building 24, CC Montpellier, France
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Gordon Langsley
- Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France; The laboratory of Comparative Cell Biology of Apicomplexa, Medical Faculty of Paris-Descartes University, Sorbonne Paris City, France.
| |
Collapse
|
163
|
Autophagy inhibitor chloroquine increases sensitivity to cisplatin in QBC939 cholangiocarcinoma cells by mitochondrial ROS. PLoS One 2017; 12:e0173712. [PMID: 28301876 PMCID: PMC5354635 DOI: 10.1371/journal.pone.0173712] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/24/2017] [Indexed: 12/21/2022] Open
Abstract
The tumor cells have some metabolic characteristics of the original tissues, and the metabolism of the tumor cells is closely related to autophagy. However, the mechanism of autophagy and metabolism in chemotherapeutic drug resistance is still poorly understood. In this study, we investigated the role and mechanism of autophagy and glucose metabolism in chemotherapeutic drug resistance by using cholangiocarcinoma QBC939 cells with primary cisplatin resistance and hepatocellular carcinoma HepG2 cells. We found that QBC939 cells with cisplatin resistance had a higher capacity for glucose uptake, consumption, and lactic acid generation, and higher activity of the pentose phosphate pathway compared with HepG2 cells, and the activity of PPP was further increased after cisplatin treatment in QBC939 cells. It is suggested that there are some differences in the metabolism of glucose in hepatocellular carcinoma and cholangiocarcinoma cells, and the activation of PPP pathway may be related to the drug resistance. Through the detection of autophagy substrates p62 and LC3, found that QBC939 cells have a higher flow of autophagy, autophagy inhibitor chloroquine can significantly increase the sensitivity of cisplatin in cholangiocarcinoma cells compared with hepatocellular carcinoma HepG2 cells. The mechanism may be related to the inhibition of QBC939 cells with higher activity of the PPP, the key enzyme G6PDH, which reduces the antioxidant capacity of cells and increases intracellular ROS, especially mitochondrial ROS. Therefore, we hypothesized that autophagy and the oxidative stress resistance mediated by glucose metabolism may be one of the causes of cisplatin resistance in cholangiocarcinoma cells. It is suggested that according to the metabolism characteristics of tumor cells, inhibition of autophagy lysosome pathway with chloroquine may be a new route for therapeutic agents against cholangiocarcinoma.
Collapse
|
164
|
Zhang YY, Gong JP, Li ZM. Autophagy and hepatic lipid metabolism. Shijie Huaren Xiaohua Zazhi 2017; 25:491-497. [DOI: 10.11569/wcjd.v25.i6.491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autophagy is initially thought to be a non-selective process in which intracellular proteins or damaged organelles are degraded. It is activated when cells lack nutrients and energy. Autophagy degrades cytoplasmic components within lysosomes and reuses the energy of amino acids to promote cell survival and maintain the cytoplasmic content. Current evidence implicates autophagy in the regulation of lipid stores within the two main organs involved in maintaining lipid homeostasis, the liver and adipose tissue. Upregulation of autophagy may lead to conversion of white adipose tissue into brown adipose tissue, thus regulating energy expenditure and obesity. Discovering new therapeutic interventions to treat lipid and lipoprotein disorders is of great interest and the discovery of autophagy as a regulator of lipid metabolism has opened up a new avenue for this area. In the liver, autophagy can play a role in some common metabolic disorders, which needs further research.
Collapse
|
165
|
White CR, Datta G, Giordano S. High-Density Lipoprotein Regulation of Mitochondrial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:407-429. [PMID: 28551800 DOI: 10.1007/978-3-319-55330-6_22] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipoproteins play a key role in regulating plasma and tissue levels of cholesterol. Apolipoprotein B (apoB)-containing lipoproteins, including chylomicrons, very-low density lipoprotein (VLDL) and low-density lipoprotein (LDL), serve as carriers of triglycerides and cholesterol and deliver these metabolites to peripheral tissues. In contrast, high-density lipoprotein (HDL) mediates Reverse Cholesterol Transport (RCT), a process by which excess cholesterol is removed from the periphery and taken up by hepatocytes where it is metabolized and excreted. Anti-atherogenic properties of HDL have been largely ascribed to apoA-I, the major protein component of the lipoprotein particle. The inflammatory response associated with atherosclerosis and ischemia-reperfusion (I-R) injury has been linked to the development of mitochondrial dysfunction. Under these conditions, an increase in reactive oxygen species (ROS) formation induces damage to mitochondrial structural elements, leading to a reduction in ATP synthesis and initiation of the apoptotic program. Recent studies suggest that HDL-associated apoA-I and lysosphingolipids attenuate mitochondrial injury by multiple mechanisms, including the suppression of ROS formation and induction of autophagy. Other apolipoproteins, however, present in lower abundance in HDL particles may exert opposing effects on mitochondrial function. This chapter examines the role of HDL-associated apolipoproteins and lipids in the regulation of mitochondrial function and bioenergetics.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Geeta Datta
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samantha Giordano
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
166
|
Yim NH, Hwang YH, Liang C, Ma JY. A platycoside-rich fraction from the root of Platycodon grandiflorum enhances cell death in A549 human lung carcinoma cells via mainly AMPK/mTOR/AKT signal-mediated autophagy induction. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:1060-1068. [PMID: 27989873 DOI: 10.1016/j.jep.2016.10.078] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of Platycodon grandiflorum (PG), commonly known as Kilkyong in Korea, Jiegeng in China, and Kikyo in Japan, has been extensively used as a traditional anti-inflammatory medicine in Asia for the treatment of respiratory conditions, such as bronchitis, asthma, and tonsillitis. Platycosides isolated from PG are especially well-known for their anti-cancer effects. AIM OF THE STUDY We investigated the involvement of autophagic cell death and other potential molecular mechanisms induced by the platycoside-containing butanol fraction of PG (PGB) in human lung carcinoma cells. MATERIALS AND METHODS PGB-induced growth inhibition and cell death were measured using a 5-diphenyl-tetrazolium bromide (MTT) assay. The effects of PGB on autophagy were determined by observing microtubule-associated protein 1 light chain 3 (LC3) redistribution with confocal microscopy. The PGB-mediated regulation of autophagy-associated proteins was investigated using Western blotting analysis. Furthermore, the anti-cancer mechanism of PGB was confirmed using chemical inhibitors. A high-performance liquid chromatography (HPLC)-DAD system was used to analyze the platycosides in PGB. RESULTS In A549 cells, PGB induced significant autophagic cell death. Specifically, PGB upregulated LC3-II in a time- and dose-dependent manner, and it redistributed LC3 via autophagosome formation in the cytoplasm. PGB treatment increased the phosphorylation of AMP-activated protein kinase (AMPK) and subsequently suppressed the AKT/mammalian target of the rapamycin (mTOR) pathway. Furthermore, PGB inhibited cell proliferation by regulating the mitogen-activated protein kinase (MAPK) pathways. In this study, six types of platycosides were identified in the PGB using HPLC. CONCLUSIONS PGB efficiently induced cancer cell death via autophagy and the modulation of the AMPK/mTOR/AKT and MAPK signaling pathways in A549 cells. Therefore, PGB may be an efficacious herbal anti-cancer therapy.
Collapse
Affiliation(s)
- Nam-Hui Yim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701-300, Republic of Korea
| | - Youn-Hwan Hwang
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701-300, Republic of Korea
| | - Chun Liang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiangshi, Henansheng 453-100, China
| | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701-300, Republic of Korea.
| |
Collapse
|
167
|
Wang Y, Huang T, Xie L, Liu L. Integrative analysis of methylation and transcriptional profiles to predict aging and construct aging specific cross-tissue networks. BMC SYSTEMS BIOLOGY 2016; 10:132. [PMID: 28155676 PMCID: PMC5260078 DOI: 10.1186/s12918-016-0354-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Aging is a complex process relating multi-scale omics data. Finding key age markers in normal tissues could help to provide reliable aging predictions in human. However, predicting age based on multi-omics data with both accuracy and informative biological function has not been performed systematically, thus relative cross-tissue analysis has not been investigated entirely, either. RESULTS Here we have developed an improved prediction pipeline, the Integrating and Stepwise Age-Prediction (ISAP) method, to regress age and find key aging markers effectively. Furthermore, we have performed a serious of network analyses, such as the PPI network, cross-tissue networks and pathway interaction networks. CONCLUSION Our results find important coordinated aging patterns between different tissues. Both co-profiling and cross-pathway analyses identify more thorough functions of aging, and could help to find aging markers, pathways and relative aging disease researches.
Collapse
Affiliation(s)
- Yin Wang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Lu Xie
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
| | - Lei Liu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China.
| |
Collapse
|
168
|
Affiliation(s)
- Ursula Matte
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Brazil
- Post-graduation Program on Genetics and Molecular Biology
- Genetics Department, Universidade Federal do Rio Grande do Sul, Brazil
| | - Gabriela Pasqualim
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Brazil
- Post-graduation Program on Genetics and Molecular Biology
- Genetics Department, Universidade Federal do Rio Grande do Sul, Brazil
| |
Collapse
|
169
|
Dos Anjos DO, Sobral Alves ES, Gonçalves VT, Fontes SS, Nogueira ML, Suarez-Fontes AM, Neves da Costa JB, Rios-Santos F, Vannier-Santos MA. Effects of a novel β-lapachone derivative on Trypanosoma cruzi: Parasite death involving apoptosis, autophagy and necrosis. Int J Parasitol Drugs Drug Resist 2016; 6:207-219. [PMID: 27770751 PMCID: PMC5078628 DOI: 10.1016/j.ijpddr.2016.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 12/18/2022]
Abstract
Natural products comprise valuable sources for new antiparasitic drugs. Here we tested the effects of a novel β-lapachone derivative on Trypanosoma cruzi parasite survival and proliferation and used microscopy and cytometry techniques to approach the mechanism(s) underlying parasite death. The selectivity index determination indicate that the compound trypanocidal activity was over ten-fold more cytotoxic to epimastigotes than to macrophages or splenocytes. Scanning electron microscopy analysis revealed that the R72 β-lapachone derivative affected the T. cruzi morphology and surface topography. General plasma membrane waving and blebbing particularly on the cytostome region were observed in the R72-treated parasites. Transmission electron microscopy observations confirmed the surface damage at the cytostome opening vicinity. We also observed ultrastructural evidence of the autophagic mechanism termed macroautophagy. Some of the autophagosomes involved large portions of the parasite cytoplasm and their fusion/confluence may lead to necrotic parasite death. The remarkably enhanced frequency of autophagy triggering was confirmed by quantitating monodansylcadaverine labeling. Some cells displayed evidence of chromatin pycnosis and nuclear fragmentation were detected. This latter phenomenon was also indicated by DAPI staining of R72-treated cells. The apoptotis induction was suggested to take place in circa one-third of the parasites assessed by annexin V labeling measured by flow cytometry. TUNEL staining corroborated the apoptosis induction. Propidium iodide labeling indicate that at least 10% of the R72-treated parasites suffered necrosis within 24 h. The present data indicate that the β-lapachone derivative R72 selectively triggers T. cruzi cell death, involving both apoptosis and autophagy-induced necrosis.
Collapse
Affiliation(s)
- Danielle Oliveira Dos Anjos
- Lab. Biologia Parasitária, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz - FIOCRUZ, Brazil; Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz UESC, Brazil
| | | | | | - Sheila Suarez Fontes
- Lab. Biologia Parasitária, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz - FIOCRUZ, Brazil
| | - Mateus Lima Nogueira
- Lab. Biologia Parasitária, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz - FIOCRUZ, Brazil
| | | | | | | | | |
Collapse
|
170
|
Redmann M, Benavides GA, Berryhill TF, Wani WY, Ouyang X, Johnson MS, Ravi S, Barnes S, Darley-Usmar VM, Zhang J. Inhibition of autophagy with bafilomycin and chloroquine decreases mitochondrial quality and bioenergetic function in primary neurons. Redox Biol 2016; 11:73-81. [PMID: 27889640 PMCID: PMC5124357 DOI: 10.1016/j.redox.2016.11.004] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 12/20/2022] Open
Abstract
Autophagy is an important cell recycling program responsible for the clearance of damaged or long-lived proteins and organelles. Pharmacological modulators of this pathway have been extensively utilized in a wide range of basic research and pre-clinical studies. Bafilomycin A1 and chloroquine are commonly used compounds that inhibit autophagy by targeting the lysosomes but through distinct mechanisms. Since it is now clear that mitochondrial quality control, particularly in neurons, is dependent on autophagy, it is important to determine whether these compounds modify cellular bioenergetics. To address this, we cultured primary rat cortical neurons from E18 embryos and used the Seahorse XF96 analyzer and a targeted metabolomics approach to measure the effects of bafilomycin A1 and chloroquine on bioenergetics and metabolism. We found that both bafilomycin and chloroquine could significantly increase the autophagosome marker LC3-II and inhibit key parameters of mitochondrial function, and increase mtDNA damage. Furthermore, we observed significant alterations in TCA cycle intermediates, particularly those downstream of citrate synthase and those linked to glutaminolysis. Taken together, these data demonstrate a significant impact of bafilomycin and chloroquine on cellular bioenergetics and metabolism consistent with decreased mitochondrial quality associated with inhibition of autophagy. Autophagy inhibition decreased mitochondrial bioenergetics in intact neurons. Autophagy inhibition decreased mitochondrial complexes I, II or IV substrate linked respiration. Autophagy inhibition increased mitochondrial DNA damage. Autophagy inhibition decreased major components of the Krebs cycle. Autophagy inhibition resulted in decreased citrate synthase activities.
Collapse
Affiliation(s)
- Matthew Redmann
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Gloria A Benavides
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Taylor F Berryhill
- Targeted Metabolomics & Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Willayat Y Wani
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Xiaosen Ouyang
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Michelle S Johnson
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Saranya Ravi
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Stephen Barnes
- Targeted Metabolomics & Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Victor M Darley-Usmar
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jianhua Zhang
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; VA Medical Center, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
171
|
Molina-Jijón E, Aparicio-Trejo OE, Rodríguez-Muñoz R, León-Contreras JC, Del Carmen Cárdenas-Aguayo M, Medina-Campos ON, Tapia E, Sánchez-Lozada LG, Hernández-Pando R, Reyes JL, Arreola-Mendoza L, Pedraza-Chaverri J. The nephroprotection exerted by curcumin in maleate-induced renal damage is associated with decreased mitochondrial fission and autophagy. Biofactors 2016; 42:686-702. [PMID: 27412471 DOI: 10.1002/biof.1313] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/26/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
Abstract
We have previously reported that the antioxidant curcumin exerts nephroprotection in maleate-induced renal damage, a model associated with oxidative stress. However, the mechanisms involved in curcumin protective effect were not explored, to assess this issue, curcumin was administered daily by gavage (150 mg/kg) five days before a single maleate (400 mg/kg)-injection. Curcumin prevented maleate-induced proteinuria, increased heat shock protein of 72 KDa (Hsp72) expression, and decreased plasma glutathione peroxidase activity. Maleate-induced oxidative stress by increasing the nicotinamide-adenine dinucleotide phosphate oxidase 4 (NOX4) and mitochondrial complex I-dependent superoxide anion (O2 •- ) production, formation of malondialdehyde (MDA)- and 3-nitrotyrosine (3-NT)-protein adducts and protein carbonylation and decreased GSH/GSSG ratio. Curcumin treatment ameliorated all the above-described changes. The maleate-induced epithelial damage, evaluated by claudin-2 and occludin expressions, was ameliorated by curcumin. It was found that maleate-induced oxidative stress promoted mitochondrial fission, evaluated by dynamin-related protein (Drp) 1 and fission (Fis) 1 expressions and by electron-microscopy, and autophagy, evaluated by phospho-threonine 389 from p70 ribosomal protein S6 kinase (p-Thr 389 p70S6K), beclin 1, microtubule-associated protein 1A/1B-light chain 3 phosphatidylethanolamine conjugate (LC3-II), autophagy-related gene 5 and 12 (Atg5-Atg12) complex, p62, and lysosomal-associated membrane protein (LAMP)-2 expressions in isolated proximal tubules and by electron-microscopy and LC-3 immunolabelling. Curcumin treatment ameliorated these changes. Moreover, curcumin alone induced autophagy in proximal tubules. These data suggest that the nephroprotective effect exerted by curcumin in maleate-induced renal damage is associated with decreased mitochondrial fission and autophagy. © 2016 BioFactors, 42(6):686-702, 2016.
Collapse
Affiliation(s)
- Eduardo Molina-Jijón
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
- Department of Biociences and Engineering, CIIEMAD-IPN, Mexico City, Mexico
| | - Omar Emiliano Aparicio-Trejo
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Rafael Rodríguez-Muñoz
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico
| | - Juan Carlos León-Contreras
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition "Salvador Zubirán" (INCMNSZ), Tlalpan, Mexico City, Mexico
| | | | - Omar Noel Medina-Campos
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Edilia Tapia
- Department of Nephrology and Laboratory of Renal Physiopathology, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Laura Gabriela Sánchez-Lozada
- Department of Nephrology and Laboratory of Renal Physiopathology, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition "Salvador Zubirán" (INCMNSZ), Tlalpan, Mexico City, Mexico
| | - José L Reyes
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico
| | | | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| |
Collapse
|
172
|
Wende AR, Young ME, Chatham J, Zhang J, Rajasekaran NS, Darley-Usmar VM. Redox biology and the interface between bioenergetics, autophagy and circadian control of metabolism. Free Radic Biol Med 2016; 100:94-107. [PMID: 27242268 PMCID: PMC5124549 DOI: 10.1016/j.freeradbiomed.2016.05.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 12/01/2022]
Abstract
Understanding molecular mechanisms that underlie the recent emergence of metabolic diseases such as diabetes and heart failure has revealed the need for a multi-disciplinary research integrating the key metabolic pathways which change the susceptibility to environmental or pathologic stress. At the physiological level these include the circadian control of metabolism which aligns metabolism with temporal demand. The mitochondria play an important role in integrating the redox signals and metabolic flux in response to the changing activities associated with chronobiology, exercise and diet. At the molecular level this involves dynamic post-translational modifications regulating transcription, metabolism and autophagy. In this review we will discuss different examples of mechanisms which link these processes together. An important pathway capable of linking signaling to metabolism is the post-translational modification of proteins by O-linked N-acetylglucosamine (O-GlcNAc). This is a nutrient regulated protein modification that plays an important role in impaired cellular stress responses. Circadian clocks have also emerged as critical regulators of numerous cardiometabolic processes, including glucose/lipid homeostasis, hormone secretion, redox status and cardiovascular function. Central to these pathways are the response of autophagy, bioenergetics to oxidative stress, regulated by Keap1/Nrf2 and mechanisms of metabolic control. The extension of these ideas to the emerging concept of bioenergetic health will be discussed.
Collapse
Affiliation(s)
- Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin E Young
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John Chatham
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namakkal S Rajasekaran
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor M Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; UAB Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
173
|
Sex-specific pharmacological modulation of autophagic process in human umbilical artery smooth muscle cells. Pharmacol Res 2016; 113:166-174. [DOI: 10.1016/j.phrs.2016.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/28/2016] [Accepted: 08/09/2016] [Indexed: 01/29/2023]
|
174
|
Mankowski RT, Ahmed S, Beaver T, Dirain M, Han C, Hess P, Martin T, Smith BK, Someya S, Leeuwenburgh C, Martin AD. Intraoperative hemidiaphragm electrical stimulation reduces oxidative stress and upregulates autophagy in surgery patients undergoing mechanical ventilation: exploratory study. J Transl Med 2016; 14:305. [PMID: 27784315 PMCID: PMC5080720 DOI: 10.1186/s12967-016-1060-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/11/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mechanical ventilation (MV) during a cardio-thoracic surgery contributes to diaphragm muscle dysfunction that impairs weaning and can lead to the ventilator- induced diaphragm dysfunction. Especially, it is critical in older adults who have lower muscle reparative capacity following MV. Reports have shown that the intraoperative intermittent hemidiaphragm electrical stimulation can maintain and/or improve post-surgery diaphragm function. In particular, from a molecular point of view, intermittent ES may reduce oxidative stress and increase regulatory autophagy levels, and therefore improve diaphragm function in animal studies. We have recently shown in humans that intraoperative ES attenuates mitochondrial dysfunction and force decline in single diaphragm muscle fibers. The aim of this study was to investigate an effect of ES on oxidative stress, antioxidant status and autophagy biomarker levels in the human diaphragm during surgery. METHODS One phrenic nerve was simulated with an external cardiac pacer in operated older subjects (62.4 ± 12.9 years) (n = 8) during the surgery. The patients received 30 pulses per min every 30 min. The muscle biopsy was collected from both hemidiaphragms and frozen for further analyses. 4-hydroxynonenal (4-HNE), an oxidative stress marker, and autophagy marker levels (Beclin-1 and the ratio of microtubule-associated protein light chain 3, I and II-LC3 II/I) protein concentrations were detected by the western blot technique. Antioxidant enzymatic activity copper-zinc (CuZnSOD) and manganese (MnSOD) superoxide dismutase were analyzed. RESULTS Levels of lipid peroxidation (4-HNE) were significantly lower in the stimulated side (p < 0.05). The antioxidant enzyme activities (CuZnSOD and MnSOD) in the stimulated side of the diaphragm were not different than in the unstimulated side (p > 0.05). Additionally, the protein concentrations of Beclin-1 and the LC3 II/I ratio were higher in the stimulated side (p < 0.05). CONCLUSION These results suggest that the intraoperative electrical stimulation decreases oxidative stress levels and upregulates autophagy levels in the stimulated hemidiaphragm. These results may contribute future studies and clinical applications on reducing post-operative diaphragm dysfunction.
Collapse
Affiliation(s)
- Robert T Mankowski
- Department of Aging and Geriatric Research, University of Florida, P.O.Box. 100107, Gainesville, FL, 32611, USA
| | - Shakeel Ahmed
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, FL, 32610-0154, USA
| | - Thomas Beaver
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Marvin Dirain
- Department of Aging and Geriatric Research, University of Florida, P.O.Box. 100107, Gainesville, FL, 32611, USA
| | - Chul Han
- Department of Aging and Geriatric Research, University of Florida, P.O.Box. 100107, Gainesville, FL, 32611, USA
| | - Phillip Hess
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Tomas Martin
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Barbara K Smith
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, FL, 32610-0154, USA
| | - Shinichi Someya
- Department of Aging and Geriatric Research, University of Florida, P.O.Box. 100107, Gainesville, FL, 32611, USA
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, University of Florida, P.O.Box. 100107, Gainesville, FL, 32611, USA.
| | - A Daniel Martin
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, FL, 32610-0154, USA.
| |
Collapse
|
175
|
Chechushkov A, Zaitseva N, Vorontsova E, Kozhin P, Menshchikova E, Shkurupiy V. Dextran loading protects macrophages from lipid peroxidation and induces a Keap1/Nrf2/ARE-dependent antioxidant response. Life Sci 2016; 166:100-107. [PMID: 27746187 DOI: 10.1016/j.lfs.2016.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/28/2016] [Accepted: 10/12/2016] [Indexed: 01/13/2023]
Abstract
AIMS Linear dextrans are often proposed as drug delivery systems with milder adverse effects and lower effective drug concentrations. Linear dextrans are polysaccharides that can potentially be used to load macrophages with drugs to transport them to a site of inflammation. Recently, it was reported that dextrans may exert a protective effect vis-à-vis drug cytotoxicity and during wound healing. The aim of the current work was to evaluate molecular mechanisms of action of dextrans that may be relevant to the cytoprotective effects. MAIN METHODS We determined the effect of treatment with 40- or 70-kDa dextran on production of reactive oxygen species, lipid peroxidation, and lysosomal pH in the J774 macrophage cell line. In addition, induction of Keap1/Nrf2/ARE and autophagic activity were evaluated. KEY FINDINGS Dextrans of both molecular weights protected the cells from oxidative stress induced by cumene hydroperoxide and from lysosomal stress induced by ammonium chloride. The effect was associated with induction of the Keap1/Nrf2/ARE signaling pathway. Furthermore, dextran stimulated autophagy in a dose-dependent manner but inhibited the autophagosome-lysosome fusion in a time-dependent manner. SIGNIFICANCE This study shows possible cytoprotective effects of dextran under oxidative stress, and these findings may be used for the development of novel (dextran-based) drug delivery approaches.
Collapse
Affiliation(s)
- Anton Chechushkov
- Research Institute of Experimental and Clinical Medicine, Novosibirsk, Russia.
| | - Natalia Zaitseva
- Research Institute of Experimental and Clinical Medicine, Novosibirsk, Russia
| | - Elena Vorontsova
- Institute of Molecular Biology and Biophysics, Novosibirsk, Russia
| | - Petr Kozhin
- Research Institute of Experimental and Clinical Medicine, Novosibirsk, Russia
| | - Elena Menshchikova
- Research Institute of Experimental and Clinical Medicine, Novosibirsk, Russia
| | - Vyacheslav Shkurupiy
- Research Institute of Experimental and Clinical Medicine, Novosibirsk, Russia; Novosibirsk State Medical University, Novosibirsk, Russia
| |
Collapse
|
176
|
Coelho RG, Cazarin JDM, Cavalcanti de Albuquerque JPA, de Andrade BM, Carvalho DP. Differential glycolytic profile and Warburg effect in papillary thyroid carcinoma cell lines. Oncol Rep 2016; 36:3673-3681. [PMID: 27748844 DOI: 10.3892/or.2016.5142] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/09/2016] [Indexed: 11/05/2022] Open
Abstract
Acceleration of glycolysis is a characteristic of neoplasia. Previous studies have shown that a metabolic shift occurs in many tumors and correlates with a negative prognosis. The present study aimed to investigate the glycolytic profile of thyroid carcinoma cell lines. We investigated glycolytic and oxidative parameters of two thyroid carcinoma papillary cell lines (BCPAP and TPC1) and the non-tumor cell line NTHY-ori. All carcinoma cell lines showed higher rates of glycolysis efficiency, when compared to NTHY-ori, as assessed by a higher rate of glucose consumption and lactate production. The BCPAP cell line presented higher rates of growth, as well as elevated intracellular ATP levels, compared to the TPC1 and NTHY-ori cells. We found that glycolysis and activities of pentose phosphate pathway (PPP) regulatory enzymes were significantly different among the carcinoma cell lines, particularly in the mitochondrial hexokinase (HK) activity which was higher in the BCPAP cells than that in the TPC1 cell line which showed a balanced distribution of HK activity between cytoplasmic and mitochondrial subcellular localizations. However, TPC1 had higher levels of glucose‑6-phosphate dehydrogenase activity, suggesting that the PPP is elevated in this cell type. Using high resolution respirometry, we observed that the Warburg effect was present in the BCPAP and TPC1 cells, characterized by low oxygen consumption and high reactive oxygen species production. Overall, these results indicate that both thyroid papillary carcinoma cell lines showed a glycolytic profile. Of note, BCPAP cells presented some relevant differences in cell metabolism compared to TPC1 cells, mainly related to higher mitochondrial-associated HK activity.
Collapse
Affiliation(s)
- Raquel Guimarães Coelho
- Laboratory of Endocrine Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Juliana de Menezes Cazarin
- Laboratory of Endocrine Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | | | - Bruno Moulin de Andrade
- Laboratory of Endocrine Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Denise P Carvalho
- Laboratory of Endocrine Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
177
|
Chen Z, Liu X, Ma S. The Roles of Mitochondria in Autophagic Cell Death. Cancer Biother Radiopharm 2016; 31:269-276. [PMID: 27754749 DOI: 10.1089/cbr.2016.2057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Zongyan Chen
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, China
- Department of Diagnostic Imaging, Center for Radiological Research, Weihai Chest Hospital, Weihai, China
| | - Xiaodong Liu
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, China
- Department of Radiation Oncology, Center for Radiological Research, The 2nd Hospital Affiliated to Jilin University, Changchun, China
| | - Shumei Ma
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
178
|
Iftinca M, Flynn R, Basso L, Melo H, Aboushousha R, Taylor L, Altier C. The stress protein heat shock cognate 70 (Hsc70) inhibits the Transient Receptor Potential Vanilloid type 1 (TRPV1) channel. Mol Pain 2016; 12:12/0/1744806916663945. [PMID: 27558883 PMCID: PMC5006304 DOI: 10.1177/1744806916663945] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/11/2016] [Indexed: 01/02/2023] Open
Abstract
Background Specialized cellular defense mechanisms prevent damage from chemical, biological, and physical hazards. The heat shock proteins have been recognized as key chaperones that maintain cell survival against a variety of exogenous and endogenous stress signals including noxious temperature. However, the role of heat shock proteins in nociception remains poorly understood. We carried out an expression analysis of the constitutively expressed 70 kDa heat-shock cognate protein, a member of the stress-induced HSP70 family in lumbar dorsal root ganglia from a mouse model of Complete Freund’s Adjuvant-induced chronic inflammatory pain. We used immunolabeling of dorsal root ganglion neurons, behavioral analysis and patch clamp electrophysiology in both dorsal root ganglion neurons and HEK cells transfected with Hsc70 and Transient Receptor Potential Channels to examine their functional interaction in heat shock stress condition. Results We report an increase in protein levels of Hsc70 in mouse dorsal root ganglia, 3 days post Complete Freund’s Adjuvant injection in the hind paw. Immunostaining of Hsc70 was observed in most of the dorsal root ganglion neurons, including the small size nociceptors immunoreactive to the TRPV1 channel. Standard whole-cell patch-clamp technique was used to record Transient Receptor Potential Vanilloid type 1 current after exposure to heat shock. We found that capsaicin-evoked currents are inhibited by heat shock in dorsal root ganglion neurons and transfected HEK cells expressing Hsc70 and TRPV1. Blocking Hsc70 with matrine or spergualin compounds prevented heat shock-induced inhibition of the channel. We also found that, in contrast to TRPV1, both the cold sensor channels TRPA1 and TRPM8 were unresponsive to heat shock stress. Finally, we show that inhibition of TRPV1 depends on the ATPase activity of Hsc70 and involves the rho-associated protein kinase. Conclusions Our work identified Hsc70 and its ATPase activity as a central cofactor of TRPV1 channel function and points to the role of this stress protein in pain associated with neurodegenerative and/or metabolic disorders, including aging.
Collapse
Affiliation(s)
- Mircea Iftinca
- Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Robyn Flynn
- Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Lilian Basso
- Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Helvira Melo
- Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Reem Aboushousha
- Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Lauren Taylor
- Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
179
|
Multiple Roles of the Small GTPase Rab7. Cells 2016; 5:cells5030034. [PMID: 27548222 PMCID: PMC5040976 DOI: 10.3390/cells5030034] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/16/2022] Open
Abstract
Rab7 is a small GTPase that belongs to the Rab family and controls transport to late endocytic compartments such as late endosomes and lysosomes. The mechanism of action of Rab7 in the late endocytic pathway has been extensively studied. Rab7 is fundamental for lysosomal biogenesis, positioning and functions, and for trafficking and degradation of several signaling receptors, thus also having implications on signal transduction. Several Rab7 interacting proteins have being identified leading to the discovery of a number of different important functions, beside its established role in endocytosis. Furthermore, Rab7 has specific functions in neurons. This review highlights and discusses the role and the importance of Rab7 on different cellular pathways and processes.
Collapse
|
180
|
Linher-Melville K, Nashed MG, Ungard RG, Haftchenary S, Rosa DA, Gunning PT, Singh G. Chronic Inhibition of STAT3/STAT5 in Treatment-Resistant Human Breast Cancer Cell Subtypes: Convergence on the ROS/SUMO Pathway and Its Effects on xCT Expression and System xc- Activity. PLoS One 2016; 11:e0161202. [PMID: 27513743 PMCID: PMC4981357 DOI: 10.1371/journal.pone.0161202] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/01/2016] [Indexed: 12/15/2022] Open
Abstract
Pharmacologically targeting activated STAT3 and/or STAT5 has been an active area of cancer research. The cystine/glutamate antiporter, system xc-, contributes to redox balance and export of intracellularly produced glutamate in response to up-regulated glutaminolysis in cancer cells. We have previously shown that blocking STAT3/5 using the small molecule inhibitor, SH-4-54, which targets the SH2 domains of both proteins, increases xCT expression, thereby increasing system xc- activity in human breast cancer cells. The current investigation demonstrates that chronic SH-4-54 administration, followed by clonal selection of treatment-resistant MDA-MB-231 and T47D breast cancer cells, elicits distinct subtype-dependent effects. xCT mRNA and protein levels, glutamate release, and cystine uptake are decreased relative to untreated passage-matched controls in triple-negative MDA-MB-231 cells, with the inverse occurring in estrogen-responsive T47D cells. This “ying-yang” effect is linked with a shifted balance between the phosphorylation status of STAT3 and STAT5, intracellular ROS levels, and STAT5 SUMOylation/de-SUMOylation. STAT5 emerged as a definitive negative regulator of xCT at the transcriptional level, while STAT3 activation is coupled with increased system xc- activity. We propose that careful classification of a patient’s breast cancer subtype is central to effectively targeting STAT3/5 as a therapeutic means of treating breast cancer, particularly given that xCT is emerging as an important biomarker of aggressive cancers.
Collapse
Affiliation(s)
- Katja Linher-Melville
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Mina G. Nashed
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Robert G. Ungard
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Sina Haftchenary
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - David A. Rosa
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Patrick T. Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- * E-mail:
| |
Collapse
|
181
|
Nourani MR, Mahmoodzadeh Hosseini H, Azimzadeh Jamalkandi S, Imani Fooladi AA. Cellular and molecular mechanisms of acute exposure to sulfur mustard: a systematic review. J Recept Signal Transduct Res 2016; 37:200-216. [DOI: 10.1080/10799893.2016.1212374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Mohammad Reza Nourani
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
182
|
Aggarwal S, Mannam P, Zhang J. Differential regulation of autophagy and mitophagy in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2016; 311:L433-52. [PMID: 27402690 PMCID: PMC5504426 DOI: 10.1152/ajplung.00128.2016] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/01/2016] [Indexed: 12/26/2022] Open
Abstract
Lysosomal-mediated degradation of intracellular lipids, proteins and organelles, known as autophagy, represents a inducible adaptive response to lung injury resulting from exposure to insults, such as hypoxia, microbes, inflammation, ischemia-reperfusion, pharmaceuticals (e.g., bleomycin), or inhaled xenobiotics (i.e., air pollution, cigarette smoke). This process clears damaged or toxic cellular constituents and facilitates cell survival in stressful environments. Autophagic degradation of dysfunctional or damaged mitochondria is termed mitophagy. Enhanced mitophagy is usually an early response to promote survival. However, overwhelming or prolonged mitochondrial damage can induce excessive/pathological levels of mitophagy, thereby promoting cell death and tissue injury. Autophagy/mitophagy is therefore an important modulator in human pulmonary diseases and a potential therapeutic target. This review article will summarize the most recent studies highlighting the role of autophagy/mitophagy and its molecular pathways involved in stress response in pulmonary pathologies.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Praveen Mannam
- Department of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
183
|
Cordani M, Oppici E, Dando I, Butturini E, Dalla Pozza E, Nadal-Serrano M, Oliver J, Roca P, Mariotto S, Cellini B, Blandino G, Palmieri M, Di Agostino S, Donadelli M. Mutant p53 proteins counteract autophagic mechanism sensitizing cancer cells to mTOR inhibition. Mol Oncol 2016; 10:1008-29. [PMID: 27118659 PMCID: PMC5423176 DOI: 10.1016/j.molonc.2016.04.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/01/2016] [Accepted: 04/04/2016] [Indexed: 11/23/2022] Open
Abstract
Mutations in TP53 gene play a pivotal role in tumorigenesis and cancer development. Here, we report that gain-of-function mutant p53 proteins inhibit the autophagic pathway favoring antiapoptotic effects as well as proliferation of pancreas and breast cancer cells. We found that mutant p53 significantly counteracts the formation of autophagic vesicles and their fusion with lysosomes throughout the repression of some key autophagy-related proteins and enzymes as BECN1 (and P-BECN1), DRAM1, ATG12, SESN1/2 and P-AMPK with the concomitant stimulation of mTOR signaling. As a paradigm of this mechanism, we show that atg12 gene repression was mediated by the recruitment of the p50 NF-κB/mutant p53 protein complex onto the atg12 promoter. Either mutant p53 or p50 NF-κB depletion downregulates atg12 gene expression. We further correlated the low expression levels of autophagic genes (atg12, becn1, sesn1, and dram1) with a reduced relapse free survival (RFS) and distant metastasis free survival (DMFS) of breast cancer patients carrying TP53 gene mutations conferring a prognostic value to this mutant p53-and autophagy-related signature. Interestingly, the mutant p53-driven mTOR stimulation sensitized cancer cells to the treatment with the mTOR inhibitor everolimus. All these results reveal a novel mechanism through which mutant p53 proteins promote cancer cell proliferation with the concomitant inhibition of autophagy.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Elisa Oppici
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Ilaria Dando
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Elena Butturini
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Elisa Dalla Pozza
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Mercedes Nadal-Serrano
- Multidisciplinar Group of Translational Oncology, University Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, E07122 Palma de Mallorca, Balearic Islands, Spain; Physiopathology of Obesity and Nutrition, CIBERobn (CB06/03), Carlos III Health Research Institute, Spain
| | - Jordi Oliver
- Multidisciplinar Group of Translational Oncology, University Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, E07122 Palma de Mallorca, Balearic Islands, Spain; Physiopathology of Obesity and Nutrition, CIBERobn (CB06/03), Carlos III Health Research Institute, Spain
| | - Pilar Roca
- Multidisciplinar Group of Translational Oncology, University Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, E07122 Palma de Mallorca, Balearic Islands, Spain; Physiopathology of Obesity and Nutrition, CIBERobn (CB06/03), Carlos III Health Research Institute, Spain
| | - Sofia Mariotto
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Barbara Cellini
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Giovanni Blandino
- Translational Oncogenomic Unit, Regina Elena National Cancer Institute-IFO, Rome, Italy
| | - Marta Palmieri
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Silvia Di Agostino
- Translational Oncogenomic Unit, Regina Elena National Cancer Institute-IFO, Rome, Italy.
| | - Massimo Donadelli
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy.
| |
Collapse
|
184
|
Basisty N, Dai D, Gagnidze A, Gitari L, Fredrickson J, Maina Y, Beyer RP, Emond MJ, Hsieh EJ, MacCoss MJ, Martin GM, Rabinovitch PS. Mitochondrial-targeted catalase is good for the old mouse proteome, but not for the young: 'reverse' antagonistic pleiotropy? Aging Cell 2016; 15:634-45. [PMID: 27061426 PMCID: PMC4933659 DOI: 10.1111/acel.12472] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2016] [Indexed: 12/31/2022] Open
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen‐containing molecules associated with aging and a broad spectrum of pathologies. We have previously shown that transgenic expression of the antioxidant enzyme catalase targeted to the mitochondria (mCAT) in mice reduces ROS, attenuates age‐related disease, and increases lifespan. However, it has been increasingly recognized that ROS also has beneficial roles in signaling, hormesis, stress response, and immunity. We therefore hypothesized that mCAT might be beneficial only when ROS approaches pathological levels in older age and might not be advantageous at a younger age when basal ROS is low. We analyzed abundance and turnover of the global proteome in hearts and livers of young (4 month) and old (20 month) mCAT and wild‐type (WT) mice. In old hearts and livers of WT mice, protein half‐lives were reduced compared to young, while in mCAT mice the reverse was observed; the longest half‐lives were seen in old mCAT mice and the shortest in young mCAT. Protein abundance of old mCAT hearts recapitulated a more youthful proteomic expression profile (P‐value < 0.01). However, young mCAT mice partially phenocopied the older wild‐type proteome (P‐value < 0.01). Age strongly interacts with mCAT, consistent with antagonistic pleiotropy in the reverse of the typical direction. These findings underscore the contrasting roles of ROS in young vs. old mice and indicate the need for better understanding of the interaction between dose and age in assessing the efficacy of therapeutic interventions in aging, including mitochondrial antioxidants.
Collapse
Affiliation(s)
- Nathan Basisty
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Dao‐Fu Dai
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Arni Gagnidze
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Lemuel Gitari
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Jeanne Fredrickson
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Yvonne Maina
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Richard P. Beyer
- Department of Environmental Health University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Mary J. Emond
- Department of Biostatistics University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Edward J. Hsieh
- Department of Genome Sciences University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Michael J. MacCoss
- Department of Genome Sciences University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - George M. Martin
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| | - Peter S. Rabinovitch
- Department of Pathology University of Washington 1959 NE Pacific Ave Seattle WA 98195 USA
| |
Collapse
|
185
|
Chang JC, Wu SL, Hoel F, Cheng YS, Liu KH, Hsieh M, Hoel A, Tronstad KJ, Yan KC, Hsieh CL, Lin WY, Kuo SJ, Su SL, Liu CS. Far-infrared radiation protects viability in a cell model of Spinocerebellar Ataxia by preventing polyQ protein accumulation and improving mitochondrial function. Sci Rep 2016; 6:30436. [PMID: 27469193 PMCID: PMC4965738 DOI: 10.1038/srep30436] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 07/05/2016] [Indexed: 01/02/2023] Open
Abstract
Far infrared radiation (FIR) is currently investigated as a potential therapeutic strategy in various diseases though the mechanism is unknown. Presently, we tested if FIR mediates beneficial effects in a cell model of the neurodegenerative disease spinocerebellar ataxia type 3 (SCA3). SCA3 is caused by a mutation leading to an abnormal polyglutamine expansion (PolyQ) in ataxin-3 protein. The consequent aggregation of mutant ataxin-3 results in disruption of vital cell functions. In this study, neuroblastoma cells (SK-N-SH) was transduced to express either non-pathogenic ataxin-3-26Q or pathogenic ataxin-3-78Q proteins. The cells expressing ataxin-3-78Q demonstrated decreased viability, and increased sensitivity to metabolic stress in the presence rotenone, an inhibitor of mitochondrial respiration. FIR exposure was found to protect against these effects. Moreover, FIR improved mitochondrial respiratory function, which was significantly compromised in ataxin-3-78Q and ataxin-3-26Q expressing cells. This was accompanied by decreased levels of mitochondrial fragmentation in FIR treated cells, as observed by fluorescence microscopy and protein expression analysis. Finally, the expression profile LC3-II, Beclin-1 and p62 suggested that FIR prevent the autophagy inhibiting effects observed in ataxin-3-78Q expressing cells. In summary, our results suggest that FIR have rescuing effects in cells expressing mutated pathogenic ataxin-3, through recovery of mitochondrial function and autophagy.
Collapse
Affiliation(s)
- Jui-Chih Chang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua 50094, Taiwan
| | - Shey-Lin Wu
- Department of Neurology, Changhua Christian Hospital, Changhua 50094, Taiwan
| | - Fredrik Hoel
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway
| | - Yu-Shan Cheng
- Department of Neurology, Changhua Christian Hospital, Changhua 50094, Taiwan
| | - Ko-Hung Liu
- Department of Neurology, Changhua Christian Hospital, Changhua 50094, Taiwan
| | - Mingli Hsieh
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan
| | - August Hoel
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway
| | | | - Kuo-Chia Yan
- Department of Dermatology, Changhua Christian Hospital, Changhua 50094, Taiwan
| | - Ching-Liang Hsieh
- Department of Chinese Medicine, Obstetrics and Gynecology, Dermatology, and Urology, China Medical University Hospital, Taichung 40447, Taiwan.,School of Chinese Medicine, Graduate Institute of Integrated Medicine, Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung 40447, Taiwan
| | - Wei-Yong Lin
- School of Chinese Medicine, Graduate Institute of Integrated Medicine, Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung 40447, Taiwan.,Departments of Medical Research, Obstetrics and Gynecology, Dermatology, and Urology, China Medical University Hospital, Taichung 40447, Taiwan
| | - Shou-Jen Kuo
- Department of Surgery, Changhua Christian Hospital, Changhua 50094, Taiwan
| | - Shih-Li Su
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Changhua Christian Hospital, Changhua 50094, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chin-San Liu
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua 50094, Taiwan.,Department of Neurology, Changhua Christian Hospital, Changhua 50094, Taiwan.,School of Chinese Medicine, Graduate Institute of Integrated Medicine, Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung 40447, Taiwan
| |
Collapse
|
186
|
Hjelmeland A, Zhang J. Metabolic, autophagic, and mitophagic activities in cancer initiation and progression. Biomed J 2016; 39:98-106. [PMID: 27372165 PMCID: PMC5514543 DOI: 10.1016/j.bj.2015.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022] Open
Abstract
Cancer is a complex disease marked by uncontrolled cell growth and invasion. These processes are driven by the accumulation of genetic and epigenetic alterations that promote cancer initiation and progression. Contributing to genome changes are the regulation of oxidative stress and reactive species-induced damage to molecules and organelles. Redox regulation, metabolic plasticity, autophagy, and mitophagy play important and interactive roles in cancer hallmarks including sustained proliferation, activated invasion, and replicative immortality. However, the impact of these processes can differ depending on the signaling pathways altered in cancer, tumor type, tumor stage, and/or the differentiation state. Here, we highlight some of the representative studies on the impact of oxidative and nitrosative activities, mitochondrial bioenergetics, metabolism, and autophagy and mitophagy in the context of tumorigenesis. We discuss the implications of these processes for cellular activities in cancer for anti-cancer-based therapeutics.
Collapse
Affiliation(s)
- Anita Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Veterans Affairs, Birmingham VA Medical Center, Birmingham, AL, USA.
| |
Collapse
|
187
|
Demers-Lamarche J, Guillebaud G, Tlili M, Todkar K, Bélanger N, Grondin M, Nguyen AP, Michel J, Germain M. Loss of Mitochondrial Function Impairs Lysosomes. J Biol Chem 2016; 291:10263-76. [PMID: 26987902 PMCID: PMC4858975 DOI: 10.1074/jbc.m115.695825] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 03/04/2016] [Indexed: 02/04/2023] Open
Abstract
Alterations in mitochondrial function, as observed in neurodegenerative diseases, lead to disrupted energy metabolism and production of damaging reactive oxygen species. Here, we demonstrate that mitochondrial dysfunction also disrupts the structure and function of lysosomes, the main degradation and recycling organelle. Specifically, inhibition of mitochondrial function, following deletion of the mitochondrial protein AIF, OPA1, or PINK1, as well as chemical inhibition of the electron transport chain, impaired lysosomal activity and caused the appearance of large lysosomal vacuoles. Importantly, our results show that lysosomal impairment is dependent on reactive oxygen species. Given that alterations in both mitochondrial function and lysosomal activity are key features of neurodegenerative diseases, this work provides important insights into the etiology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Julie Demers-Lamarche
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| | - Gérald Guillebaud
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| | - Mouna Tlili
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Kiran Todkar
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| | - Noémie Bélanger
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Martine Grondin
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Angela P Nguyen
- the Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Jennifer Michel
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Marc Germain
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| |
Collapse
|
188
|
Iizumi T, Takahashi S, Mashima K, Minami K, Izawa Y, Abe T, Hishiki T, Suematsu M, Kajimura M, Suzuki N. A possible role of microglia-derived nitric oxide by lipopolysaccharide in activation of astroglial pentose-phosphate pathway via the Keap1/Nrf2 system. J Neuroinflammation 2016; 13:99. [PMID: 27143001 PMCID: PMC4855896 DOI: 10.1186/s12974-016-0564-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/26/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) plays a pivotal role in the pathophysiology of stroke-induced inflammation. Both astroglia and microglia express TLR4, and endogenous ligands produced in the ischemic brain induce inflammatory responses. Reactive oxygen species (ROS), nitric oxide (NO), and inflammatory cytokines produced by TLR4 activation play harmful roles in neuronal damage after stroke. Although astroglia exhibit pro-inflammatory responses upon TLR4 stimulation by lipopolysaccharide (LPS), they may also play cytoprotective roles via the activation of the pentose phosphate pathway (PPP), reducing oxidative stress by glutathione peroxidase. We investigated the mechanisms by which astroglia reduce oxidative stress via the activation of PPP, using TLR4 stimulation and hypoxia in concert with microglia. METHODS In vitro experiments were performed using cells prepared from Sprague-Dawley rats. Coexisting microglia in the astroglial culture were chemically eliminated using L-leucine methyl ester (LME). Cells were exposed to LPS (0.01 μg/mL) or hypoxia (1 % O2) for 12-15 h. PPP activity was measured using [1-(14)C]glucose and [6-(14)C]glucose. ROS and NO production were measured using 2',7'-dichlorodihydrofluorescein diacetate and diaminofluorescein-FM diacetate, respectively. The involvement of nuclear factor-erythroid-2-related factor 2 (Nrf2), a cardinal transcriptional factor under stress conditions that regulates glucose 6-phosphate dehydrogenase, the rate-limiting enzyme of PPP, was evaluated using immunohistochemistry. RESULTS Cultured astroglia exposed to LPS elicited 20 % increases in PPP flux, and these actions of astroglia appeared to involve Nrf2. However, the chemical depletion of coexisting microglia eliminated both increases in PPP and astroglial nuclear translocation of Nrf2. LPS induced ROS and NO production in the astroglial culture containing microglia but not in the microglia-depleted astroglial culture. LPS enhanced astroglial ROS production after glutathione depletion. U0126, an upstream inhibitor of mitogen-activated protein kinase, eliminated LPS-induced NO production, whereas ROS production was unaffected. U0126 also eliminated LPS-induced PPP activation in astroglial-microglial culture, indicating that microglia-derived NO mediated astroglial PPP activation. Hypoxia induced astroglial PPP activation independent of the microglia-NO pathway. Elimination of ROS and NO production by sulforaphane, a natural Nrf2 activator, confirmed the astroglial protective mechanism. CONCLUSIONS Astroglia in concert with microglia may play a cytoprotective role for countering oxidative stress in stroke.
Collapse
Affiliation(s)
- Takuya Iizumi
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Shinichi Takahashi
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| | - Kyoko Mashima
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Kazushi Minami
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yoshikane Izawa
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Takato Abe
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka-shi, 545-8585, Osaka , Japan
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,Clinical and Translational Research Center, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Mayumi Kajimura
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Norihiro Suzuki
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| |
Collapse
|
189
|
Huber-Villaume S, Revelant G, Sibille E, Philippot S, Morabito A, Dunand S, Chaimbault P, Bagrel D, Kirsch G, Hesse S, Schohn H. 2-(Thienothiazolylimino)-1,3-thiazolidin-4-ones inhibit cell division cycle 25 A phosphatase. Bioorg Med Chem 2016; 24:2920-2928. [PMID: 27178385 DOI: 10.1016/j.bmc.2016.04.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/28/2016] [Accepted: 04/30/2016] [Indexed: 11/26/2022]
Abstract
Cell division cycle dual phosphatases (CDC25) are essential enzymes that regulate cell progression in cell cycle. Three isoforms exist as CDC25A, B and C. Over-expression of each CDC25 enzyme is found in cancers of diverse origins. Thiazolidinone derivatives have been reported to display anti-proliferative activities, bactericidal activities and to reduce inflammation process. New 2-(thienothiazolylimino)-1,3-thiazolidin-4-ones were synthesized and evaluated as inhibitors of CDC25 phosphatase. Among the molecules tested, compound 6 inhibited CDC25A with an IC50 estimated at 6.2±1.0μM. The binding of thiazolidinone derivative 6 onto CDC25A protein was reversible. In cellulo, compound 6 treatment led to MCF7 and MDA-MB-231 cell growth arrest. To our knowledge, it is the first time that such 4-thiazolidinone derivatives are characterized as CDC25 potential inhibitor.
Collapse
Affiliation(s)
- Sophie Huber-Villaume
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 5 (MIC), Campus Bridoux, rue du Général Delestraint, 57070 Metz Cedex, France
| | - Germain Revelant
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 3 (HECRIN), 1 Boulevard Arago, 57078 Metz Technopôle, France
| | - Estelle Sibille
- Université de Lorraine, EA 4632-Laboratoire de Chimie et Physique Approche Multi-échelle des Milieux Complexes, 1 boulevard Arago, 57078 Metz Cedex 3, France
| | - Stéphanie Philippot
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 5 (MIC), Campus Bridoux, rue du Général Delestraint, 57070 Metz Cedex, France
| | - Angelica Morabito
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 5 (MIC), Campus Bridoux, rue du Général Delestraint, 57070 Metz Cedex, France
| | - Sandrine Dunand
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 3 (HECRIN), 1 Boulevard Arago, 57078 Metz Technopôle, France
| | - Patrick Chaimbault
- Université de Lorraine, EA 4632-Laboratoire de Chimie et Physique Approche Multi-échelle des Milieux Complexes, 1 boulevard Arago, 57078 Metz Cedex 3, France
| | - Denyse Bagrel
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 5 (MIC), Campus Bridoux, rue du Général Delestraint, 57070 Metz Cedex, France
| | - Gilbert Kirsch
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 3 (HECRIN), 1 Boulevard Arago, 57078 Metz Technopôle, France
| | - Stéphanie Hesse
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 3 (HECRIN), 1 Boulevard Arago, 57078 Metz Technopôle, France.
| | - Hervé Schohn
- Université de Lorraine, UMR CNRS 7565, Structure et Réactivité des Systèmes Moléculaires Complexes, Equipe 5 (MIC), Campus Bridoux, rue du Général Delestraint, 57070 Metz Cedex, France.
| |
Collapse
|
190
|
McVey KA, Snapp IB, Johnson MB, Negga R, Pressley AS, Fitsanakis VA. Exposure of C. elegans eggs to a glyphosate-containing herbicide leads to abnormal neuronal morphology. Neurotoxicol Teratol 2016; 55:23-31. [PMID: 27019975 PMCID: PMC4884470 DOI: 10.1016/j.ntt.2016.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 03/20/2016] [Accepted: 03/20/2016] [Indexed: 12/21/2022]
Abstract
Recent data demonstrate that chronic exposure of Caenorhabditis elegans (C. elegans) to a high-use glyphosate-containing herbicide, Touchdown (TD), potentially damages the adult nervous system. It is unknown, however, whether unhatched worms exposed to TD during the egg stage show abnormal neurodevelopment post-hatching. Therefore, we investigated whether early treatment with TD leads to aberrant neuronal or neurite development in C. elegans. Studies were completed in three different worm strains with green fluorescent protein (GFP)-tagged neurons to facilitate visual neuronal assessment. Initially, eggs from C. elegans with all neurons tagged with GFP were chronically exposed to TD. Visual inspection suggested decreased neurite projections associated with ventral nerve cord neurons. Data analysis showed a statistically significant decrease in overall green pixel numbers at the fourth larval (L4) stage (*p<0.05). We further investigated whether specific neuronal populations were preferentially vulnerable to TD by treating eggs from worms that had all dopaminergic (DAergic) or γ-aminobutyric acid (GABAergic) neurons tagged with GFP. As before, green pixel number associated with these discrete neuronal populations was analyzed at multiple larval stages. Data analysis indicated statistically significant decreases in pixel number associated with DAergic, but not GABAergic, neurons (***p<0.001) at all larval stages. Finally, statistically significant decreases (at the first larval stage, L1) or increases (at the fourth larval stage, L4) in superoxide levels, a developmental signaling molecule, were detected (*p<0.05). These data suggest that early exposure to TD may impair neuronal development, perhaps through superoxide perturbation. Since toxic insults during development may late render individuals more vulnerable to neurodegenerative diseases in adulthood, these studies provide some of the first evidence in this model organism that early exposure to TD may adversely affect the developing nervous system.
Collapse
Affiliation(s)
- Kenneth A McVey
- King University, Department of Biology, 1350 King College Road, Bristol, TN 37620, USA; Liberty University College of Osteopathic Medicine, 306 Liberty View Lane, Lynchburg, VA 24502, USA.
| | - Isaac B Snapp
- King University, Department of Biology, 1350 King College Road, Bristol, TN 37620, USA; Medical University of South Carolina, Physician Assistant Studies, 151B Rutledge Avenue, Charleston, SC 29425, USA.
| | - Megan B Johnson
- King University, Department of Biology, 1350 King College Road, Bristol, TN 37620, USA; Lincoln Memorial University, Debusk College of Osteopathic Medicine, 6965 Cumberland Gap Parkway, Harrogate, TN 37752, USA.
| | - Rekek Negga
- King University, Department of Biology, 1350 King College Road, Bristol, TN 37620, USA; The University of Tennessee Institute of Agriculture, Department of Animal Science, 366 Brehm Animal Science Building, 2506 River Drive, Knoxville, TN 37996, USA.
| | - Aireal S Pressley
- King University, Department of Biology, 1350 King College Road, Bristol, TN 37620, USA.
| | - Vanessa A Fitsanakis
- King University, Department of Biology, 1350 King College Road, Bristol, TN 37620, USA.
| |
Collapse
|
191
|
Khoshnood B, Dacklin I, Grabbe C. Urm1: an essential regulator of JNK signaling and oxidative stress in Drosophila melanogaster. Cell Mol Life Sci 2016; 73:1939-54. [PMID: 26715182 PMCID: PMC11108535 DOI: 10.1007/s00018-015-2121-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/28/2015] [Accepted: 12/15/2015] [Indexed: 12/29/2022]
Abstract
Ubiquitin-related modifier 1 (Urm1) is a ubiquitin-like molecule (UBL) with the dual capacity to act both as a sulphur carrier and posttranslational protein modifier. Here we characterize the Drosophila melanogaster homologues of Urm1 (CG33276) and its E1 activating enzyme Uba4 (CG13090), and show that they function together to induce protein urmylation in vivo. Urm1 conjugation to target proteins in general, and to the evolutionary conserved substrate Peroxiredoxin 5 (Prx5) specifically, is dependent on Uba4. A complete loss of Urm1 is lethal in flies, although a small number of adult zygotic Urm1 (n123) mutant escapers can be recovered. These escapers display a decreased general fitness and shortened lifespan, but in contrast to their S. cerevisiae counterparts, they are resistant to oxidative stress. Providing a molecular explanation, we demonstrate that cytoprotective JNK signaling is increased in Urm1 deficient animals. In agreement, molecular and genetic evidence suggest that elevated activity of the JNK downstream target genes Jafrac1 and gstD1 strongly contributes to the tolerance against oxidative stress displayed by Urm1 (n123) null mutants. In conclusion, Urm1 is a UBL that is involved in the regulation of JNK signaling and the response against oxidative stress in the fruit fly.
Collapse
Affiliation(s)
- B Khoshnood
- Department of Molecular Biology, Umeå University, Building 6L, 901 87, Umeå, Sweden
| | - I Dacklin
- Department of Molecular Biology, Umeå University, Building 6L, 901 87, Umeå, Sweden
| | - C Grabbe
- Department of Molecular Biology, Umeå University, Building 6L, 901 87, Umeå, Sweden.
| |
Collapse
|
192
|
Ye F, Kaneko H, Hayashi Y, Takayama K, Hwang SJ, Nishizawa Y, Kimoto R, Nagasaka Y, Tsunekawa T, Matsuura T, Yasukawa T, Kondo T, Terasaki H. Malondialdehyde induces autophagy dysfunction and VEGF secretion in the retinal pigment epithelium in age-related macular degeneration. Free Radic Biol Med 2016; 94:121-34. [PMID: 26923802 DOI: 10.1016/j.freeradbiomed.2016.02.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/19/2016] [Accepted: 02/22/2016] [Indexed: 11/22/2022]
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness in developed countries and is closely related to oxidative stress, which leads to lipid peroxidation. Malondialdehyde (MDA) is a major byproduct of polyunsaturated fatty acid (PUFA) peroxidation. Increased levels of MDA have been reported in eyes of AMD patients. However, little is known about the direct relationship between MDA and AMD. Here we show the biological importance of MDA in AMD pathogenesis. We first confirmed that MDA levels were significantly increased in eyes of AMD patients. In ARPE-19 cells, a human retinal pigment epithelial cell line, MDA treatment induced vascular endothelial growth factor (VEGF) expression alternation, cell junction disruption, and autophagy dysfunction that was also observed in eyes of AMD patients. The MDA-induced VEGF increase was inhibited by autophagy-lysosomal inhibitors. Intravitreal MDA injection in mice increased laser-induced choroidal neovascularization (laser-CNV) volumes. In a mouse model fed a high-linoleic acid diet for 3 months, we found a significant increase in MDA levels, autophagic activity, and laser-CNV volumes. Our study revealed an important role of MDA, which acts not only as a marker but also as a causative factor of AMD pathogenesis-related autophagy dysfunction. Furthermore, higher dietary intake of linoleic acid promoted CNV progression in mice with increased MDA levels.
Collapse
Affiliation(s)
- Fuxiang Ye
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hiroki Kaneko
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Yumi Hayashi
- Department of Radiological and Medical Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan; Institute for Advanced Research, Nagoya University, Nagoya 464-8601, Japan
| | - Kei Takayama
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shiang-Jyi Hwang
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Laboratory of Bell Research Center-Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yuji Nishizawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Reona Kimoto
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yosuke Nagasaka
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Taichi Tsunekawa
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Toshiyuki Matsuura
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tsutomu Yasukawa
- Department of Ophthalmology, Nagoya City University School of Medicine, Nagoya 467-8601, Japan
| | - Takaaki Kondo
- Department of Radiological and Medical Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Hiroko Terasaki
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
193
|
Wang X, Bai Y, Cheng G, Ihsan A, Zhu F, Wang Y, Tao Y, Chen D, Dai M, Liu Z, Yuan Z. Genomic and proteomic analysis of the inhibition of synthesis and secretion of aldosterone hormone induced by quinocetone in NCI-H295R cells. Toxicology 2016; 350-352:1-14. [PMID: 27046791 DOI: 10.1016/j.tox.2016.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 11/26/2022]
Abstract
Quinoxaline 1,4-dioxides (QdNOs) are widely used as a kind of antibacterial growth promoter in animal husbandry. The adrenal cortex was found to be one of the main toxic targets of QdNOs, accompanied by a decreased aldosterone level. However, the way in which QdNOs decrease production of the hormone aldosterone is far from clear. To illustrate the mechanism by which QdNOs damage the adrenal cortex and decrease aldosterone hormone levels, the QdNOs were screened to choose the drug with most toxic effects on aldosterone production, and then to reveal the mechanism between the gene and protein profiles in human adrenocortical cells (NCI-H295R cells). The results found that quinocetone (QCT) showed the highest adrenal toxic effect among QdNOs. After exposing H295R cells to 10 and 20μM QCT for 24h, compared with blank cells, the gene and protein expression profiles obtained were analyzed by microarray and MALDI TOF/TOF mass spectrometry, respectively. The results of microarray analysis suggested that ABCG1 and SREBF1, which were involved in the cholesterol biosynthetic and metabolic processes, and CYP17A1, NR4A2 and G6PD, which were related to aldosterone biosynthesis, were important molecular targets. It has been speculated that PKC and ERK pathways might be involved in the reduction of aldosterone production caused by QCT, through enhanced mRNA expression of CYP17A1. Additionally, JNK and p38MAPK signal transduction pathways might participate in apoptosis induced by QCT. Twenty-nine and 32 protein spots were successfully identified when cells were treated with 10 and 20μM QCT, respectively. These identified proteins mainly included material synthesis and energy metabolism-related proteins, transcription/translation processing-related proteins, signal transduction proteins, cytoskeletal proteins, molecular chaperones, proteins related to response to stress, and transport proteins. Further investigations suggested that oxidative stress caused by QCT was exacerbated through disruption of the Keap1/Nrf2/ARE anti-oxidative stress pathway. Taken together, the data demonstrated for the first time that the Keap1/Nrf2/ARE pathway plays a crucial role in adrenal toxicity, and that CYP17A1 was the key switch to reduce the aldosterone production induced by QCT. Furthermore, large numbers of genes and proteins and entry points for research in the inhibition of aldosterone synthesis induced by QCT were offered, which will provide new insight into the adrenal toxicity of QdNOs and help to provide a theoretical foundation for the formulation of safety controls for products obtained from animals and to design new QdNOs with less harmful effects.
Collapse
Affiliation(s)
- Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yijie Bai
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Guyue Cheng
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Awais Ihsan
- Department of Biosciences, COMSATS Institute of Information Technology, Sahiwal, Pakistan
| | - Feng Zhu
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yulian Wang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yanfei Tao
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Dongmei Chen
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Menghong Dai
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Zhengli Liu
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China
| | - Zonghui Yuan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, Hubei, China.
| |
Collapse
|
194
|
Unravelling the relationship between macroautophagy and mitochondrial ROS in cancer therapy. Apoptosis 2016; 21:517-31. [DOI: 10.1007/s10495-016-1236-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
195
|
Li P, Zhang D, Shen L, Dong K, Wu M, Ou Z, Shi D. Redox homeostasis protects mitochondria through accelerating ROS conversion to enhance hypoxia resistance in cancer cells. Sci Rep 2016; 6:22831. [PMID: 26956544 PMCID: PMC4783784 DOI: 10.1038/srep22831] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/24/2016] [Indexed: 11/09/2022] Open
Abstract
Mitochondria are the powerhouses of eukaryotic cells and the main source of reactive oxygen species (ROS) in hypoxic cells, participating in regulating redox homeostasis. The mechanism of tumor hypoxia tolerance, especially the role of mitochondria in tumor hypoxia resistance remains largely unknown. This study aimed to explore the role of mitochondria in tumor hypoxia resistance. We observed that glycolysis in hypoxic cancer cells was up-regulated more rapidly, with far lesser attenuation in aerobic oxidation, thus contributing to a more stable ATP/ADP ratio. In hypoxia, cancer cells rapidly convert hypoxia-induced O(2˙)(-) into H2O2. H2O2 is further decomposed by a relatively stronger antioxidant system, causing ROS levels to increase lesser compared to normal cells. The moderate ROS leads to an appropriate degree of autophagy, eliminating the damaged mitochondria and offering nutrients to promote mitochondria fusion, thus protects mitochondria and improves hypoxia tolerance in cancer. The functional mitochondria could enable tumor cells to flexibly switch between glycolysis and oxidative phosphorylation to meet the different physiological requirements during the hypoxia/re-oxygenation cycling of tumor growth.
Collapse
Affiliation(s)
- Pengying Li
- Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| | - Dongyang Zhang
- Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| | - Lingxiao Shen
- Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| | - Kelei Dong
- Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| | - Meiling Wu
- Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| | - Zhouluo Ou
- Cancer Research Institute of Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China
| | - Dongyun Shi
- Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
196
|
Walters JW, Amos D, Ray K, Santanam N. Mitochondrial redox status as a target for cardiovascular disease. Curr Opin Pharmacol 2016; 27:50-5. [PMID: 26894468 DOI: 10.1016/j.coph.2016.01.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/25/2016] [Accepted: 01/29/2016] [Indexed: 02/07/2023]
Abstract
Mitochondria are major players in cellular energetics, oxidative stress and programmed cell death. Mitochondrial dynamics regulate and integrate these functions. Mitochondrial dysfunction is involved in cardiac hypertrophy, hypertension and myocardial ischemia/reperfusion injury. Reactive oxygen species generation is modulated by the fusion-fission pathway as well as key proteins such as sirtuins that act as metabolic sensors of cellular energetics. Mitochondrial redox status has thus become a good target for therapy against cardiovascular diseases. Recently, there is an influx of studies garnered towards assessing the beneficial effects of mitochondrial targeted antioxidants, drugs modulating the fusion-fission proteins, sirtuins, and other mitochondrial processes as potential cardio-protecting agents.
Collapse
Affiliation(s)
- James W Walters
- School of Arts & Sciences, Bluefield State College, Basic Science Building B213, 219 Rock Street, Bluefield, WV 24701, USA
| | - Deborah Amos
- Department of Pharmacology, Physiology & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV 25755, USA
| | - Kristeena Ray
- Department of Pharmacology, Physiology & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV 25755, USA
| | - Nalini Santanam
- Department of Pharmacology, Physiology & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV 25755, USA.
| |
Collapse
|
197
|
Ursini F, Maiorino M, Forman HJ. Redox homeostasis: The Golden Mean of healthy living. Redox Biol 2016; 8:205-15. [PMID: 26820564 PMCID: PMC4732014 DOI: 10.1016/j.redox.2016.01.010] [Citation(s) in RCA: 284] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 01/15/2016] [Accepted: 01/18/2016] [Indexed: 12/20/2022] Open
Abstract
The notion that electrophiles serve as messengers in cell signaling is now widely accepted. Nonetheless, major issues restrain acceptance of redox homeostasis and redox signaling as components of maintenance of a normal physiological steady state. The first is that redox signaling requires sudden switching on of oxidant production and bypassing of antioxidant mechanisms rather than a continuous process that, like other signaling mechanisms, can be smoothly turned up or down. The second is the misperception that reactions in redox signaling involve “reactive oxygen species” rather than reaction of specific electrophiles with specific protein thiolates. The third is that hormesis provides protection against oxidants by increasing cellular defense or repair mechanisms rather than by specifically addressing the offset of redox homeostasis. Instead, we propose that both oxidant and antioxidant signaling are main features of redox homeostasis. As the redox shift is rapidly reversed by feedback reactions, homeostasis is maintained by continuous signaling for production and elimination of electrophiles and nucleophiles. Redox homeostasis, which is the maintenance of nucleophilic tone, accounts for a healthy physiological steady state. Electrophiles and nucleophiles are not intrinsically harmful or protective, and redox homeostasis is an essential feature of both the response to challenges and subsequent feedback. While the balance between oxidants and nucleophiles is preserved in redox homeostasis, oxidative stress provokes the establishment of a new radically altered redox steady state. The popular belief that scavenging free radicals by antioxidants has a beneficial effect is wishful thinking. We propose, instead, that continuous feedback preserves nucleophilic tone and that this is supported by redox active nutritional phytochemicals. These nonessential compounds, by activating Nrf2, mimic the effect of endogenously produced electrophiles (parahormesis). In summary, while hormesis, although globally protective, results in setting up of a new phenotype, parahormesis contributes to health by favoring maintenance of homeostasis. Redox homeostasis is the continuously challenged oxidative/nucleophilic balance. Rheostatic redox signaling enzymes maintain oxidative/nucleophilic homeostasis. Phytochemicals assist redox homeostasis through oxidative feedback (parahormesis). Adaptation and hormesis while protective establish a new phenotype and set point.
Collapse
Affiliation(s)
- Fulvio Ursini
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, I-35121 Padova, Italy
| | - Matilde Maiorino
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, I-35121 Padova, Italy
| | - Henry Jay Forman
- Andrus Gerontology Center of the Davis School of Gerontology, University of Southern, California, 3715 McClintock Ave, Los Angeles, CA 90089-0191, USA
| |
Collapse
|
198
|
Piedrahita D, Castro-Alvarez JF, Boudreau RL, Villegas-Lanau A, Kosik KS, Gallego-Gomez JC, Cardona-Gómez GP. β-Secretase 1's Targeting Reduces Hyperphosphorilated Tau, Implying Autophagy Actors in 3xTg-AD Mice. Front Cell Neurosci 2016; 9:498. [PMID: 26778963 PMCID: PMC4705306 DOI: 10.3389/fncel.2015.00498] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/11/2015] [Indexed: 01/08/2023] Open
Abstract
β-site APP cleaving enzyme 1 (BACE1) initiates APP cleavage, which has been reported to be an inducer of tau pathology by altering proteasome functions in Alzheimer’s disease (AD). However, the exact relationship between BACE1 and PHF (Paired Helical Filaments) formation is not clear. In this study, we confirm that BACE1 and Hsc70 are upregulated in the brains of AD patients, and we demonstrate that both proteins show enhanced expression in lipid rafts from AD-affected triple transgenic mouse brains. BACE1 targeting increased Hsc70 levels in the membrane and cytoplasm fractions and downregulated Hsp90 and CHIP in the nucleus in the hippocampi of 3xTg-AD mice. However, these observations occurred in a proteasome-independent manner in vitro. The BACE1miR-induced reduction of soluble hyperphosphorylated tau was associated with a decrease in MAPK activity. However, the BACE1 RNAi-mediated reduction of hyperphosphorylated tau was only blocked by 3-MA (3-methyladenine) in vitro, and it resulted in the increase of Hsc70 and LAMP2 in lipid rafts from hippocampi of 3xTg-AD mice, and upregulation of survival and homeostasis signaling. In summary, our findings suggest that BACE1 silencing neuroprotects reducing soluble hyperphosphorylated tau, modulating certain autophagy-related proteins in aged 3xTg-AD mice.
Collapse
Affiliation(s)
- Diego Piedrahita
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| | - John Fredy Castro-Alvarez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| | | | - Andres Villegas-Lanau
- Neurobank, Neuroscience Group of Antioquia, Faculty of Medicine, SIU, University of Antioquia Medellín, Colombia
| | - Kenneth S Kosik
- Department of Molecular Cellular Developmental Biology, Neuroscience Research Institute, University of California Santa Barbara Santa Barbara, CA, USA
| | - Juan Carlos Gallego-Gomez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| | - Gloria Patricia Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Viral Vector Core and Gene Therapy, University of Antioquia Medellin, Antioquia, Colombia
| |
Collapse
|
199
|
Rodrigues D, Viotto AC, Checchia R, Gomide A, Severino D, Itri R, Baptista MS, Martins WK. Mechanism of Aloe Vera extract protection against UVA: shelter of lysosomal membrane avoids photodamage. Photochem Photobiol Sci 2016; 15:334-50. [DOI: 10.1039/c5pp00409h] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Aloe Vera extract exhibited remarkable ability of reducing both in vitro and in vivo photodamage, even though it does not have anti-radical properties.
Collapse
Affiliation(s)
| | | | | | - Andreza Gomide
- Instituto de Física
- Universidade de São Paulo
- Brazil
- Centro Universitário Padre Anchieta
- Brazil
| | | | | | | | | |
Collapse
|
200
|
Oxidative Stress in the Healthy and Wounded Hepatocyte: A Cellular Organelles Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:8327410. [PMID: 26788252 PMCID: PMC4691634 DOI: 10.1155/2016/8327410] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 09/10/2015] [Indexed: 02/06/2023]
Abstract
Accurate control of the cell redox state is mandatory for maintaining the structural integrity and physiological functions. This control is achieved both by a fine-tuned balance between prooxidant and anti-oxidant molecules and by spatial and temporal confinement of the oxidative species. The diverse cellular compartments each, although structurally and functionally related, actively maintain their own redox balance, which is necessary to fulfill specialized tasks. Many fundamental cellular processes such as insulin signaling, cell proliferation and differentiation and cell migration and adhesion, rely on localized changes in the redox state of signal transducers, which is mainly mediated by hydrogen peroxide (H2O2). Therefore, oxidative stress can also occur long before direct structural damage to cellular components, by disruption of the redox circuits that regulate the cellular organelles homeostasis. The hepatocyte is a systemic hub integrating the whole body metabolic demand, iron homeostasis and detoxification processes, all of which are redox-regulated processes. Imbalance of the hepatocyte's organelles redox homeostasis underlies virtually any liver disease and is a field of intense research activity. This review recapitulates the evolving concept of oxidative stress in the diverse cellular compartments, highlighting the principle mechanisms of oxidative stress occurring in the healthy and wounded hepatocyte.
Collapse
|