1
|
Almeida F, Ferreira IL, Naia L, Marinho D, Vilaça-Ferreira AC, Costa MD, Duarte-Silva S, Maciel P, Rego AC. Mitochondrial Dysfunction and Decreased Cytochrome c in Cell and Animal Models of Machado-Joseph Disease. Cells 2023; 12:2397. [PMID: 37830611 PMCID: PMC10571982 DOI: 10.3390/cells12192397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023] Open
Abstract
Mitochondrial dysfunction has been described in many neurodegenerative disorders; however, there is less information regarding mitochondrial deficits in Machado-Joseph disease (MJD), a polyglutamine (polyQ) disorder caused by CAG repeat expansion in the ATXN3 gene. In the present study, we characterized the changes in mitochondrial function and biogenesis markers in two MJD models, CMVMJD135 (MJD135) transgenic mice at a fully established phenotype stage and tetracycline-regulated PC6-3 Q108 cell line expressing mutant ataxin-3 (mATXN3). We detected mATXN3 in the mitochondrial fractions of PC6-3 Q108 cells, suggesting the interaction of expanded ATXN3 with the organelle. Interestingly, in both the cerebella of the MJD135 mouse model and in PC6-3 Q108 cells, we found decreased mitochondrial respiration, ATP production and mitochondrial membrane potential, strongly suggesting mitochondrial dysfunction in MJD. Also, in PC6-3 Q108 cells, an additional enhanced glycolytic flux was observed. Supporting the functional deficits observed in MJD mitochondria, MJD135 mouse cerebellum and PC6-3 Q108 cells showed reduced cytochrome c mRNA and protein levels. Overall, our findings show compromised mitochondrial function associated with decreased cytochrome c levels in both cell and animal models of MJD.
Collapse
Affiliation(s)
- Filipa Almeida
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (F.A.); (I.L.F.); (L.N.); (D.M.)
| | - Ildete L. Ferreira
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (F.A.); (I.L.F.); (L.N.); (D.M.)
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Luana Naia
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (F.A.); (I.L.F.); (L.N.); (D.M.)
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Daniela Marinho
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (F.A.); (I.L.F.); (L.N.); (D.M.)
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Ana Catarina Vilaça-Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.C.V.-F.); (M.D.C.); (S.D.-S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Marta D. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.C.V.-F.); (M.D.C.); (S.D.-S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.C.V.-F.); (M.D.C.); (S.D.-S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.C.V.-F.); (M.D.C.); (S.D.-S.)
- ICVS/3B’s-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - A. Cristina Rego
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (F.A.); (I.L.F.); (L.N.); (D.M.)
- FMUC-Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
2
|
Chang H, Hu X, Tang X, Tian S, Li Y, Lv X, Shang L. A Mitochondria-Targeted Fluorescent Probe for Monitoring NADPH Overproduction during Influenza Virus Infection. ACS Sens 2023; 8:829-838. [PMID: 36689687 DOI: 10.1021/acssensors.2c02458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Reduced nicotinamide adenine dinucleotide phosphate (NADPH) is an important cofactor in the progress of antioxidant synthesis and biosynthesis, and an abnormal NADPH level has been observed in many viral infection processes. However, efficient tools to monitor NADPH in living cells after viral infection have not been reported. In this work, we present a fluorescent probe, NAFP4, that could detect NADPH ex vivo with a low detection limit of 3.66 nM and image mitochondrial NADPH level changes in living cells. The probe exhibits excellent cell permeability, rapid reactivity, and high selectivity with minimal cytotoxicity. Using NAFP4, we reveal that the NADPH is overproduced in the host cells infected by influenza virus, which was caused by an elevated level of G6PDH during the virus infection. Moreover, there was positive association between the G6PDH level and virus replication. With the proposed probe NAFP4, our study highlights that the virus infection would influence the host metabolism in NADPH production and also suggests that G6PDH is expected to be a promising target for antiviral therapy.
Collapse
Affiliation(s)
- Hao Chang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, No. 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China.,Drug Discovery Center for Infectious Disease, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Xiao Hu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, No. 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China.,Drug Discovery Center for Infectious Disease, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Xiaomei Tang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, No. 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China.,Drug Discovery Center for Infectious Disease, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Shiwei Tian
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, No. 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China.,Drug Discovery Center for Infectious Disease, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Yidan Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, No. 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China.,Drug Discovery Center for Infectious Disease, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Xing Lv
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, No. 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China.,Drug Discovery Center for Infectious Disease, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| | - Luqing Shang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and KLMDASR of Tianjin, Nankai University, No. 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China.,Drug Discovery Center for Infectious Disease, Nankai University, 38 Tongyan Road, Haihe Education Park, Tianjin 300350, People's Republic of China
| |
Collapse
|
3
|
Möller M, Orrico F, Villar S, López AC, Silva N, Donzé M, Thomson L, Denicola A. Oxidants and Antioxidants in the Redox Biochemistry of Human Red Blood Cells. ACS OMEGA 2023; 8:147-168. [PMID: 36643550 PMCID: PMC9835686 DOI: 10.1021/acsomega.2c06768] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/09/2022] [Indexed: 06/01/2023]
Abstract
Red blood cells (RBCs) are exposed to both external and internal sources of oxidants that challenge their integrity and compromise their physiological function and supply of oxygen to tissues. Autoxidation of oxyhemoglobin is the main source of endogenous RBC oxidant production, yielding superoxide radical and then hydrogen peroxide. In addition, potent oxidants from other blood cells and the surrounding endothelium can reach the RBCs. Abundant and efficient enzymatic systems and low molecular weight antioxidants prevent most of the damage to the RBCs and also position the RBCs as a sink of vascular oxidants that allow the body to maintain a healthy circulatory system. Among the antioxidant enzymes, the thiol-dependent peroxidase peroxiredoxin 2, highly abundant in RBCs, is essential to keep the redox balance. A great part of the RBC antioxidant activity is supported by an active glucose metabolism that provides reducing power in the form of NADPH via the pentose phosphate pathway. There are several RBC defects and situations that generate oxidative stress conditions where the defense mechanisms are overwhelmed, and these include glucose-6-phosphate dehydrogenase deficiencies (favism), hemoglobinopathies like sickle cell disease and thalassemia, as well as packed RBCs for transfusion that suffer from storage lesions. These oxidative stress-associated pathologies of the RBCs underline the relevance of redox balance in these anucleated cells that lack a mechanism of DNA-inducible antioxidant response and rely on a complex and robust network of antioxidant systems.
Collapse
Affiliation(s)
- Matias
N. Möller
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Florencia Orrico
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Sebastián
F. Villar
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Ana C. López
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Nicolás Silva
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
- Departamento
de Medicina Transfusional, Hospital de Clínicas, Facultad de
Medicina, Universidad de la República, Montevideo 11600, Uruguay
| | - Marcel Donzé
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Leonor Thomson
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Ana Denicola
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| |
Collapse
|
4
|
Ju S, Chen H, Wang S, Lin J, Ma Y, Aroian RV, Peng D, Sun M. C. elegans monitor energy status via the AMPK pathway to trigger innate immune responses against bacterial pathogens. Commun Biol 2022; 5:643. [PMID: 35773333 PMCID: PMC9246835 DOI: 10.1038/s42003-022-03589-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Pathogen recognition and the triggering of host innate immune system are critical to understanding pathogen-host interaction. Cellular surveillance systems have been identified as an important strategy for the identification of microbial infection. In the present study, using Bacillus thuringiensis-Caenorhabditis elegans as a model, we found an approach for surveillance systems to sense pathogens. We report that Bacillus thuringiensis Cry5Ba, a typical pore-forming toxin, caused mitochondrial damage and energy imbalance by triggering potassium ion leakage, instead of directly targeting mitochondria. Interestingly, we find C. elegans can monitor intracellular energy status to trigger innate immune responses via AMP-activated protein kinase (AMPK), secreting multiple effectors to defend against pathogenic attacks. Our study indicates that the imbalance of energy status is a prevalent side effect of pathogen infection. Furthermore, the AMPK-dependent surveillance system may serve as a practicable strategy for the host to recognize and defense against pathogens. Bacillus thuringiensis toxin Cry5Ba triggers potassium ion leakage, causing mitochondrial damage and energy imbalance. C. elegans can monitor this intracellular energy imbalance via AMP-activated protein kinase to trigger innate immune responses.
Collapse
Affiliation(s)
- Shouyong Ju
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hanqiao Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shaoying Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Lin
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yanli Ma
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Raffi V Aroian
- Program in Molecular Medicine, University of Massachusetts Chan Medical School Worcester, Worcester, MA, 01605-2377, USA
| | - Donghai Peng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Ming Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
5
|
Giner MP, Christen S, Bartova S, Makarov MV, Migaud ME, Canto C, Moco S. A Method to Monitor the NAD + Metabolome-From Mechanistic to Clinical Applications. Int J Mol Sci 2021; 22:10598. [PMID: 34638936 PMCID: PMC8508997 DOI: 10.3390/ijms221910598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 01/07/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) and its reduced form (NADH) are coenzymes employed in hundreds of metabolic reactions. NAD+ also serves as a substrate for enzymes such as sirtuins, poly(ADP-ribose) polymerases (PARPs) and ADP-ribosyl cyclases. Given the pivotal role of NAD(H) in health and disease, studying NAD+ metabolism has become essential to monitor genetic- and/or drug-induced perturbations related to metabolic status and diseases (such as ageing, cancer or obesity), and its possible therapies. Here, we present a strategy based on liquid chromatography-tandem mass spectrometry (LC-MS/MS), for the analysis of the NAD+ metabolome in biological samples. In this method, hydrophilic interaction chromatography (HILIC) was used to separate a total of 18 metabolites belonging to pathways leading to NAD+ biosynthesis, including precursors, intermediates and catabolites. As redox cofactors are known for their instability, a sample preparation procedure was developed to handle a variety of biological matrices: cell models, rodent tissues and biofluids, as well as human biofluids (urine, plasma, serum, whole blood). For clinical applications, quantitative LC-MS/MS for a subset of metabolites was demonstrated for the analysis of the human whole blood of nine volunteers. Using this developed workflow, our methodology allows studying NAD+ biology from mechanistic to clinical applications.
Collapse
Affiliation(s)
- Maria Pilar Giner
- Nestle Research, EPFL Innovation Park, H, 1015 Lausanne, Switzerland; (M.P.G.); (S.C.); (S.B.); (C.C.)
| | - Stefan Christen
- Nestle Research, EPFL Innovation Park, H, 1015 Lausanne, Switzerland; (M.P.G.); (S.C.); (S.B.); (C.C.)
| | - Simona Bartova
- Nestle Research, EPFL Innovation Park, H, 1015 Lausanne, Switzerland; (M.P.G.); (S.C.); (S.B.); (C.C.)
| | - Mikhail V. Makarov
- Mitchell Cancer Institute, University of South Alabama, 1660 Springhill Avenue, Mobile, AL 36604, USA; (M.V.M.); (M.E.M.)
- Olon Ricerca Bioscience, 7528 Auburn Road, Concord, OH 44077, USA
| | - Marie E. Migaud
- Mitchell Cancer Institute, University of South Alabama, 1660 Springhill Avenue, Mobile, AL 36604, USA; (M.V.M.); (M.E.M.)
| | - Carles Canto
- Nestle Research, EPFL Innovation Park, H, 1015 Lausanne, Switzerland; (M.P.G.); (S.C.); (S.B.); (C.C.)
| | - Sofia Moco
- Nestle Research, EPFL Innovation Park, H, 1015 Lausanne, Switzerland; (M.P.G.); (S.C.); (S.B.); (C.C.)
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
6
|
Sex-dependent vulnerability of fetal nonhuman primate cardiac mitochondria to moderate maternal nutrient reduction. Clin Sci (Lond) 2021; 135:1103-1126. [PMID: 33899910 DOI: 10.1042/cs20201339] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
Poor maternal nutrition in pregnancy affects fetal development, predisposing offspring to cardiometabolic diseases. The role of mitochondria during fetal development on later-life cardiac dysfunction caused by maternal nutrient reduction (MNR) remains unexplored. We hypothesized that MNR during gestation causes fetal cardiac bioenergetic deficits, compromising cardiac mitochondrial metabolism and reserve capacity. To enable human translation, we developed a primate baboon model (Papio spp.) of moderate MNR in which mothers receive 70% of control nutrition during pregnancy, resulting in intrauterine growth restriction (IUGR) offspring and later exhibiting myocardial remodeling and heart failure at human equivalent ∼25 years. Term control and MNR baboon offspring were necropsied following cesarean-section, and left ventricle (LV) samples were collected. MNR adversely impacted fetal cardiac LV mitochondria in a sex-dependent fashion. Increased maternal plasma aspartate aminotransferase, creatine phosphokinase (CPK), and elevated cortisol levels in MNR concomitant with decreased blood insulin in male fetal MNR were measured. MNR resulted in a two-fold increase in fetal LV mitochondrial DNA (mtDNA). MNR resulted in increased transcripts for several respiratory chain (NDUFB8, UQCRC1, and cytochrome c) and adenosine triphosphate (ATP) synthase proteins. However, MNR fetal LV mitochondrial complex I and complex II/III activities were significantly decreased, possibly contributing to the 73% decreased ATP content and increased lipid peroxidation. MNR fetal LV showed mitochondria with sparse and disarranged cristae dysmorphology. Conclusion: MNR disruption of fetal cardiac mitochondrial fitness likely contributes to the documented developmental programming of adult cardiac dysfunction, indicating a programmed mitochondrial inability to deliver sufficient energy to cardiac tissues as a chronic mechanism for later-life heart failure.
Collapse
|
7
|
Guidarelli A, Cerioni L, Fiorani M, Catalani A, Cantoni O. Arsenite-Induced Mitochondrial Superoxide Formation: Time and Concentration Requirements for the Effects of the Metalloid on the Endoplasmic Reticulum and Mitochondria. J Pharmacol Exp Ther 2020; 373:62-71. [PMID: 31941719 DOI: 10.1124/jpet.119.262469] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/13/2020] [Indexed: 03/08/2025] Open
Abstract
The present study used human myeloid leukemia U937 cells, a versatile promonocytic cellular system that, based on its endoplasmic reticulum (ER)/mitochondria functional relationships, responds to low micromolar concentrations of arsenite with a single, defined mechanism of superoxide (O2 -.) formation. Under these conditions, we observe an initial Ca2+ mobilization from the ER associated with the mitochondrial accumulation of the cation, which is followed by Ca2+-dependent mitochondrial O2 -. (mitoO2 -.) formation. These events, which were barely detectable after 3 hours, were better appreciated at 6 hours. We found that markedly shorter exposure to arsenite and lower concentrations of arsenite are required to induce extensive O2 - formation in cells supplemented with inositol-1,4,5-trisphosphate receptor (IP3R) or ryanodine receptor (RyR) agonists. Indeed, nanomolar arsenite induced maximal O2 -. formation after only 10 minutes of exposure, and this response was uniquely dependent on the enforced mitochondrial Ca2+ accumulation. The dramatic anticipation of and sensitization to the effects of arsenite caused by the IP3R or RyR agonists were accompanied by a parallel significant genotoxic response in the absence of detectable mitochondrial dysfunction and cytotoxicity. We conclude that the prolonged, low-micromolar arsenite exposure paradigm resulting in mitoO2 -. formation is necessary to affect Ca2+ homeostasis and accumulate the cation in mitochondria. The arsenite requirements to promote mitoO2 -. formation in the presence of sufficient mitochondrial Ca2+ were instead remarkably lower in terms of both concentration and time of exposure. These conditions were associated with the induction of extensive DNA strand scission in the absence of detectable signs of toxicity. SIGNIFICANCE STATEMENT: In respiration-proficient cells, arsenite causes mitochondrial Ca2+ accumulation and Ca2+-dependent mitochondrial superoxide formation. We now report that the second event requires remarkably lower concentrations of and time of exposure to the metalloid than the former. Indeed, a brief exposure to nanomolar levels of arsenite produced maximal effects under conditions in which the mitochondrial Ca2+ concentration ([Ca2+]m) was increased by inositol-1,4,5-trisphosphate receptor or ryanodine receptor agonists. Hence, specific substances or conditions enhancing the [Ca2+]m may potentiate the deleterious effects of arsenite by selectively increasing mitochondrial superoxide formation.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Liana Cerioni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Alessia Catalani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| |
Collapse
|
8
|
Roch A, Magon NJ, Maire J, Suarna C, Ayer A, Waldvogel S, Imhof BA, Koury MJ, Stocker R, Schapira M. Transition to 37°C reveals importance of NADPH in mitigating oxidative stress in stored RBCs. JCI Insight 2019; 4:126376. [PMID: 31581149 DOI: 10.1172/jci.insight.126376] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 09/25/2019] [Indexed: 01/14/2023] Open
Abstract
The RBC storage lesion is a multiparametric response that occurs during storage at 4°C, but its impact on transfused patients remains unclear. In studies of the RBC storage lesion, the temperature transition from cold storage to normal body temperature that occurs during transfusion has received limited attention. We hypothesized that multiple deleterious events might occur in this period of increasing temperature. We show dramatic alterations in several properties of therapeutic blood units stored at 4°C after warming them to normal body temperature (37°C), as well as febrile temperature (40°C). In particular, the intracellular content and redox state of NADP(H) were directly affected by post-storage incubation at 37°C, as well as by pro-oxidant storage conditions. Modulation of the NADPH-producing pentose phosphate pathway, but not the prevention of hemoglobin autoxidation by conversion of oxyhemoglobin to carboxyhemoglobin, provided protection against storage-induced alterations in RBCs, demonstrating the central role of NADPH in mitigating increased susceptibility of stored RBCs to oxidative stress. We propose that assessing RBC oxidative status after restoration of body temperature constitutes a sensitive method for detecting storage-related alterations that has the potential to improve the quality of stored RBCs for transfusion.
Collapse
Affiliation(s)
- Aline Roch
- Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Nicholas J Magon
- Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Jessica Maire
- Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Cacang Suarna
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Anita Ayer
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales Medicine, Sydney, New South Wales, Australia
| | - Sophie Waldvogel
- Centre de Transfusion Sanguine, University Hospitals, University of Geneva, Geneva, Switzerland
| | - Beat A Imhof
- Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Mark J Koury
- Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales Medicine, Sydney, New South Wales, Australia
| | | |
Collapse
|
9
|
Metabolic Effects of Dietary Glycerol Supplementation in Muscle and Liver of European Seabass and Rainbow Trout by 1H NMR Metabolomics. Metabolites 2019; 9:metabo9100202. [PMID: 31569727 PMCID: PMC6835518 DOI: 10.3390/metabo9100202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 12/17/2022] Open
Abstract
The sustainable growth of fish aquaculture will require the procurement of non-marine feed sources. Glycerol is a potential feed supplement whose metabolism may spare the catabolism of dietary amino acids, thereby extending the use of the feed protein to other physiological functions such as growth. In the present study, the effects of dietary glycerol supplementation on the muscle and liver metabolomes of rainbow trout (Oncorhynchus mykiss) and European seabass (Dicentrarchus labrax) were evaluated. Fish juveniles were fed diets with 0%, 2.5%, and 5% glycerol. Muscle and liver aqueous fractions were extracted and 1H NMR spectra were acquired. Metabolite profiles derived from the 1H NMR signals were assessed using univariate and multivariate statistical analyses. The adenylate energy charge was determined in the muscle. For both species, the muscle metabolite profile showed more variability compared to that of the liver and was most perturbed by the 5.0% glycerol diet. For the liver metabolite profile, rainbow trout showed fewer differences compared to European seabass. No differences were observed in energy charge between experimental groups for either species. Thus, rainbow trout appeared to be less susceptible to tissue metabolite perturbations, compared to seabass, when the diet was supplemented with up to 5% glycerol.
Collapse
|
10
|
Martins RM, Teodoro JS, Furtado E, Rolo AP, Palmeira CM, Tralhão JG. Evaluation of bioenergetic and mitochondrial function in liver transplantation. Clin Mol Hepatol 2019; 25:190-198. [PMID: 30897898 PMCID: PMC6589847 DOI: 10.3350/cmh.2018.0087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/14/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND/AIMS We measured changes in mitochondrial function and bioenergetics that occur during ischemia/ reperfusion in fresh liver samples of patients undergoing liver transplantation. These variations correlated with markers of liver function and clinical outcome. Ischemia/reperfusion injury related to liver transplantation affects mitochondrial function and bioenergetics. Experimental studies were conducted to identify the role of bioenergetics and mitochondrial dysfunction. To the best of our knowledge, no investigation of these two factors' impacts on liver transplantation has been performed. METHODS This was a prospective study of 28 patients who underwent liver transplantation. We measured parameters of mitochondrial function and bioenergetics in biopsies performed during the procedure. RESULTS We observed a statistically significant reduction in mitochondrial membrane potential, an increase in lag phase, and decreases in mitochondrial respiration and adenosine triphosphate content (P<0.010). Higher postoperative aminotransferase peaks correlated with worse mitochondrial function; mitochondrial respiration correlated with arterial lactate (P<0.010). CONCLUSION There is a relationship between mitochondrial function and ischemia/reperfusion injury. The future use of these clinical markers as prognostic factors may allow early identification of post-transplant liver failure and may indicate the need to perform a new transplant.
Collapse
Affiliation(s)
| | - João Soeiro Teodoro
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Emanuel Furtado
- Adult and Paediatric Liver Transplantation Unit, Coimbra University and Hospital Centre, Coimbra, Portugal
| | - Anabela Pinto Rolo
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Carlos Marques Palmeira
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Guilherme Tralhão
- General Surgery Department, Coimbra University and Hospital Centre, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
11
|
Martins RM, Teodoro JS, Furtado E, Oliveira RC, Tralhão JG, Rolo AP, Palmeira CM. Mild hypothermia during the reperfusion phase protects mitochondrial bioenergetics against ischemia-reperfusion injury in an animal model of ex-vivo liver transplantation-an experimental study. Int J Med Sci 2019; 16:1304-1312. [PMID: 31588197 PMCID: PMC6775262 DOI: 10.7150/ijms.34617] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
The organ preservation paradigm has changed following the development of new ways to preserve organs. The use of machine perfusion to preserve organs appears to have several advantages compared with conventional static cold storage. For liver transplants, the temperature control provided by machine perfusion improves organ preservation. In this experimental study, we measured the effects of different temperatures on mitochondrial bioenergetics during the reperfusion phase. An experimental model of ex-vivo liver transplantation was developed in Wistar rats (Rattus norvegicus). After total hepatectomy, cold static preservation occurred at 4ºC and reperfusion was performed at 37ºC and 32ºC using a Langendorff system. We measured parameters associated with mitochondrial bioenergetics in the livers. Compared with the livers that underwent normothermic reperfusion, mild hypothermia during reperfusion caused significant increases in the mitochondrial membrane potential, the adenosine triphosphate content, and mitochondrial respiration, and a significant reduction in the lag phase (all P < 0.001). Mild hypothermia during reperfusion reduced the effect of ischemia-reperfusion injury on mitochondrial activity in liver tissue and promoted an increase in bioenergetic availability compared with normothermic reperfusion.
Collapse
Affiliation(s)
- Rui Miguel Martins
- Department of Surgery, Instituto Português de Oncologia de Coimbra, Coimbra, Portugal
| | - João Soeiro Teodoro
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra; and Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Emanuel Furtado
- Unidade de Transplantação Hepática de Crianças e Adultos, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Rui Caetano Oliveira
- Department of Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - José Guilherme Tralhão
- Department of Surgery, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Clínica Universitária de Cirurgia III, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; and Center for Investigation on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Anabela Pinto Rolo
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra; and Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Carlos Marques Palmeira
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra; and Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
12
|
Guidarelli A, Fiorani M, Cantoni O. Low Concentrations of Arsenite Target the Intraluminal Inositol 1, 4, 5-Trisphosphate Receptor/Ryanodine Receptor Crosstalk to Significantly Elevate Intracellular Ca 2. J Pharmacol Exp Ther 2018; 367:184-193. [PMID: 30068729 DOI: 10.1124/jpet.118.250480] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
Arsenite is an established human carcinogen that induces cytotoxic and genotoxic effects through poorly defined mechanisms involving the formation of reactive oxygen species (ROS) and deregulated Ca2+ homeostasis. We used variants of the U937 cell line to address the central issue of the mechanism whereby arsenite affects Ca2+ homeostasis. We found that 6-hour exposure to the metalloid (2.5 μM), although not associated with an immediate or delayed toxicity, causes a significant increase in the intracellular Ca2+ concentration ([Ca2+]i) through a mechanism characterized by the following components: 1) it was not affected by ROS produced under the same conditions; 2) a small amount of Ca2+ was mobilized from the inositol-1,4,5-trisphosphate receptor (IP3R), and this response was not augmented by greater concentrations of the metalloid; 3) large amounts of Ca2+ were instead dose dependently mobilized from the ryanodine receptor (RyR) in response to IP3R stimulation; 4) the cells maintained an intact responsiveness to agonist-stimulated Ca2+ mobilization from both channels; 5) arsenite, even at 5-10 µM, failed to directly mobilize Ca2+ from the RyR; and 6) arsenite failed to enhance Ca2+ release from the RyR under conditions in which the [Ca2+]i was increased by either RyR agonists or ionophore-stimulated Ca2+ uptake. We therefore conclude that arsenite elevates the [Ca2+]i by directly targeting the IP3R and its intraluminal crosstalk with the RyR. This mechanism likely mediates mitochondrial superoxide formation, downstream damage on various biomolecules (including genomic DNA), and mitochondrial dysfunction/apoptosis eventually occurring after longer incubation to, or exposure to greater concentrations of, arsenite.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
13
|
Monteiro C, Ferreira de Oliveira JMP, Pinho F, Bastos V, Oliveira H, Peixoto F, Santos C. Biochemical and transcriptional analyses of cadmium-induced mitochondrial dysfunction and oxidative stress in human osteoblasts. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2018; 81:705-717. [PMID: 29913117 DOI: 10.1080/15287394.2018.1485122] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cadmium (Cd) accumulation is known to occur predominantly in kidney and liver; however, low-level long-term exposure to Cd may also result in bone damage. Few studies have addressed Cd-induced toxicity in osteoblasts, particularly upon cell mitochondrial energy processing and putative associations with oxidative stress in bone. To assess the influence of Cd treatment on mitochondrial function and oxidative status in osteoblast cells, human MG-63 cells were treated with Cd (up to 65 μM) for 24 or 48 h. Intracellular reactive oxygen species (ROS), lipid and protein oxidation and antioxidant defense mechanisms such as total antioxidant activity (TAA) and gene expression of antioxidant enzymes were analyzed. In addition, Cd-induced effects on mitochondrial function were assessed by analyzing the activity of enzymes involved in mitochondrial respiration, membrane potential (ΔΨm), mitochondrial morphology and adenylate energy charge. Treatment with Cd increased oxidative stress, concomitantly with lipid and protein oxidation. Real-time polymerase chain reaction (qRT-PCR) analyses of antioxidant genes catalase (CAT), glutathione peroxidase 1 (GPX1), glutathione S-reductase (GSR), and superoxide dismutase (SOD1 and SOD2) exhibited a trend toward decrease in transcripts in Cd-stressed cells, particularly a downregulation of GSR. Longer treatment with Cd (48 h) resulted in energy charge states significantly below those commonly observed in living cells. Mitochondrial function was affected by ΔΨm reduction. Inhibition of mitochondrial respiratory chain enzymes and citrate synthase also occurred following Cd treatment. In conclusion, Cd induced mitochondrial dysfunction which appeared to be associated with oxidative stress in human osteoblasts.
Collapse
Affiliation(s)
- Cristina Monteiro
- a Department of Biology & CESAM , University of Aveiro, Campus Universitário , Aveiro , Portugal
| | - José Miguel P Ferreira de Oliveira
- b LAQV/REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy , University of Porto , Porto , Portugal
| | - Francisco Pinho
- a Department of Biology & CESAM , University of Aveiro, Campus Universitário , Aveiro , Portugal
| | - Verónica Bastos
- c Department of Biology & LAQV/REQUIMTE , Faculty of Sciences of University of Porto , Porto , Portugal
| | - Helena Oliveira
- a Department of Biology & CESAM , University of Aveiro, Campus Universitário , Aveiro , Portugal
| | - Francisco Peixoto
- d Biology and Environment Department , Chemistry Research Center, University of Trás-os-Montes & Alto Douro , Portugal
| | - Conceição Santos
- c Department of Biology & LAQV/REQUIMTE , Faculty of Sciences of University of Porto , Porto , Portugal
| |
Collapse
|
14
|
Duarte AI, Sjögren M, Santos MS, Oliveira CR, Moreira PI, Björkqvist M. Dual Therapy with Liraglutide and Ghrelin Promotes Brain and Peripheral Energy Metabolism in the R6/2 Mouse Model of Huntington's Disease. Sci Rep 2018; 8:8961. [PMID: 29895889 PMCID: PMC5997749 DOI: 10.1038/s41598-018-27121-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023] Open
Abstract
Neuronal loss alongside altered energy metabolism, are key features of Huntington’s disease (HD) pathology. The orexigenic gut-peptide hormone ghrelin is known to stimulate appetite and affect whole body energy metabolism. Liraglutide is an efficient anti-type 2 diabetes incretin drug, with neuroprotective effects alongside anorectic properties. Combining liraglutide with the orexigenic peptide ghrelin may potentially promote brain/cognitive function in HD. The R6/2 mouse model of HD exhibits progressive central pathology, weight loss, deranged glucose metabolism, skeletal muscle atrophy and altered body composition. In this study, we targeted energy metabolism in R6/2 mice using a co-administration of liraglutide and ghrelin. We investigated their effect on brain cortical hormone-mediated intracellular signalling pathways, metabolic and apoptotic markers, and the impact on motor function in HD. We here demonstrate that liraglutide, alone or together with ghrelin (subcutaneous daily injections of 150 µg/kg (ghrelin) and 0.2 mg/kg (liraglutide), for 2 weeks), normalized glucose homeostatic features in the R6/2 mouse, without substantially affecting body weight or body composition. Liraglutide alone decreased brain cortical active GLP-1 and IGF-1 levels in R6/2 mice, alongside higher ADP levels. Liraglutide plus ghrelin decreased brain insulin, lactate, AMP and cholesterol levels in R6/2 mice. Taken together, our findings encourage further studies targeting energy metabolism in HD.
Collapse
Affiliation(s)
- Ana I Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal. .,Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.
| | - Marie Sjögren
- Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Maria S Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Life Sciences Department, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Catarina R Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Biochemistry, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Björkqvist
- Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Stocchi V, Canestrari F, Giacchi R, Sebastiani M, Lungarotti F, Dachà U, Dachà M. Adenine and Pyridine Nucleotides in the Red Blood Cells of Subjects with Solid Tumors. TUMORI JOURNAL 2018; 73:25-8. [PMID: 3824530 DOI: 10.1177/030089168707300104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The concentration of adenine (ATP, ADP, AMP) and pyridine (NADP+, NADPH, NAD+, NADH) nucleotides in the erythrocytes of subjects affected by solid tumors was evaluated using a method which allows their simultaneous extraction and reverse-phase high-performance liquid chromatographic analysis. The results showed a lower level of ATP in the erythrocytes of subjects affected by solid tumors, whereas no significant modifications were observed in the other compounds. In fact, the mean value of ATP in these subjects was 27 % lower than that of normal adults. This fact is discussed in relation to other enzymatic and metabolic modifications previously observed in red blood cells.
Collapse
|
16
|
Yaku K, Okabe K, Nakagawa T. Simultaneous measurement of NAD metabolome in aged mice tissue using liquid chromatography tandem-mass spectrometry. Biomed Chromatogr 2018; 32:e4205. [PMID: 29424941 DOI: 10.1002/bmc.4205] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/08/2018] [Accepted: 01/25/2018] [Indexed: 12/19/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD) is a major co-factor that mediates multiple biological processes including redox reaction and gene expression. Recently, NAD metabolism has received considerable attention because administration of NAD precursors exhibited beneficial effects against aging-related metabolic disorders in animals. Although numerous studies have reported that NAD levels decline with aging in multiple animal tissues, the pathway and kinetics of NAD metabolism in aged organs are not completely understood. To determine the NAD metabolism upon aging, we developed targeted metabolomics based on an LC/MS/MS system. Our method is simple and applicable to crude biological samples, including culture cells and animal tissues. Unlike a conventional enzymatic cycling assay, our approach can determine NAD and NADH (reduced form of NAD) by performing a single sample preparation. Further, we validated our method using biological samples and investigated the alteration of the NAD metabolome during aging. Consistent with previous reports, the NAD levels in the liver and skeletal muscle decreased with aging. Further, we detected a significant increase in nicotinamide mononucleotide and nicotinamide riboside in the kidney upon aging. The LC/MS/MS-based NAD metabolomics that we have developed is extensively applicable to biomedical studies, and the results will present innovative ideas for the aging studies, especially for that of NAD metabolism.
Collapse
Affiliation(s)
- Keisuke Yaku
- Frontier Research Core for Life Sciences, University of Toyama, Toyama, Japan.,Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Keisuke Okabe
- Frontier Research Core for Life Sciences, University of Toyama, Toyama, Japan.,Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan.,First Department of Internal Medicine, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Takashi Nakagawa
- Frontier Research Core for Life Sciences, University of Toyama, Toyama, Japan.,Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan.,Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
17
|
Abstract
SIGNIFICANCE The nicotinamide adenine dinucleotide (NAD+)/reduced NAD+ (NADH) and NADP+/reduced NADP+ (NADPH) redox couples are essential for maintaining cellular redox homeostasis and for modulating numerous biological events, including cellular metabolism. Deficiency or imbalance of these two redox couples has been associated with many pathological disorders. Recent Advances: Newly identified biosynthetic enzymes and newly developed genetically encoded biosensors enable us to understand better how cells maintain compartmentalized NAD(H) and NADP(H) pools. The concept of redox stress (oxidative and reductive stress) reflected by changes in NAD(H)/NADP(H) has increasingly gained attention. The emerging roles of NAD+-consuming proteins in regulating cellular redox and metabolic homeostasis are active research topics. CRITICAL ISSUES The biosynthesis and distribution of cellular NAD(H) and NADP(H) are highly compartmentalized. It is critical to understand how cells maintain the steady levels of these redox couple pools to ensure their normal functions and simultaneously avoid inducing redox stress. In addition, it is essential to understand how NAD(H)- and NADP(H)-utilizing enzymes interact with other signaling pathways, such as those regulated by hypoxia-inducible factor, to maintain cellular redox homeostasis and energy metabolism. FUTURE DIRECTIONS Additional studies are needed to investigate the inter-relationships among compartmentalized NAD(H)/NADP(H) pools and how these two dinucleotide redox couples collaboratively regulate cellular redox states and cellular metabolism under normal and pathological conditions. Furthermore, recent studies suggest the utility of using pharmacological interventions or nutrient-based bioactive NAD+ precursors as therapeutic interventions for metabolic diseases. Thus, a better understanding of the cellular functions of NAD(H) and NADP(H) may facilitate efforts to address a host of pathological disorders effectively. Antioxid. Redox Signal. 28, 251-272.
Collapse
Affiliation(s)
- Wusheng Xiao
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Rui-Sheng Wang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Diane E Handy
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
18
|
Addition of Berberine to Preservation Solution in an Animal Model of Ex Vivo Liver Transplant Preserves Mitochondrial Function and Bioenergetics from the Damage Induced by Ischemia/Reperfusion. Int J Mol Sci 2018; 19:ijms19010284. [PMID: 29351246 PMCID: PMC5796230 DOI: 10.3390/ijms19010284] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/12/2018] [Accepted: 01/14/2018] [Indexed: 02/07/2023] Open
Abstract
Liver transplantation is a therapeutic regimen to treat patients with non-malignant end-stage liver diseases and malignant tumors of hepatic origin. The ischemia/reperfusion (I/R) injury in liver transplantation is associated with disruption of mitochondrial function in the hepatic parenchyma. Several studies have been conducted in animal models to identify pharmacological therapeutic strategies to minimize the injury induced by the cold/warm I/R in liver transplantation. Most of these studies were conducted in unrealistic conditions without the potential to be translated to clinical usage. Berberine (BBR) is a pharmacological compound with a potential protective effect of the mitochondrial function in the context of I/R. For the future clinical application of these pharmacological strategies, it is essential that a close resemblance exists between the methodology used in the animals models and real life. In this study, we have demonstrated that the addition of BBR to the preservation solution in an I/R setting preserves mitochondrial function and bioenergetics, protecting the liver from the deleterious effects caused by I/R. As such, BBR has the potential to be used as a pharmacological therapeutic strategy.
Collapse
|
19
|
Lee MH, Koh D, Na H, Ka NL, Kim S, Kim HJ, Hong S, Shin YK, Seong JK, Lee MO. MTA1 is a novel regulator of autophagy that induces tamoxifen resistance in breast cancer cells. Autophagy 2018; 14:812-824. [PMID: 29130361 PMCID: PMC6070012 DOI: 10.1080/15548627.2017.1388476] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 09/18/2017] [Accepted: 10/02/2017] [Indexed: 12/22/2022] Open
Abstract
Tamoxifen is commonly used to treat patients with ESR/ER-positive breast cancer, but its therapeutic benefit is limited by the development of resistance. Recently, alterations in macroautophagy/autophagy function were demonstrated to be a potential mechanism for tamoxifen resistance. Although MTA1 (metastasis-associated 1) has been implicated in breast tumorigenesis and metastasis, its role in endocrine resistance has not been studied. Here, we report that the level of MTA1 expression was upregulated in the tamoxifen resistant breast cancer cell lines MCF7/TAMR and T47D/TR, and knockdown of MTA1 sensitized the cells to 4-hydroxytamoxifen (4OHT). Moreover, knockdown of MTA1 significantly decreased the enhanced autophagy flux in the tamoxifen resistant cell lines. To confirm the role of MTA1 in the development of tamoxifen resistance, we established a cell line, MCF7/MTA1, which stably expressed MTA1. Compared with parental MCF7, MCF7/MTA1 cells were more resistant to 4OHT-induced growth inhibition in vitro and in vivo, and showed increased autophagy flux and higher numbers of autophagosomes. Knockdown of ATG7 or cotreatment with hydroxychloroquine, an autophagy inhibitor, restored sensitivity to 4OHT in both the MCF7/MTA1 and tamoxifen resistant cells. In addition, AMP-activated protein kinase (AMPK) was activated, probably because of an increased AMP:ATP ratio and decreased expression of mitochondrial electron transport complex components. Finally, publicly available breast cancer patient datasets indicate that MTA1 levels correlate with poor prognosis and development of recurrence in patients with breast cancer treated with tamoxifen. Overall, our findings demonstrated that MTA1 induces AMPK activation and subsequent autophagy that could contribute to tamoxifen resistance in breast cancer.
Collapse
Affiliation(s)
- Min-Ho Lee
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Dahae Koh
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Hyelin Na
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Na-Lee Ka
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Seungsu Kim
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Hyeon-Ji Kim
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Sungyoul Hong
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Young Kee Shin
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University and Korea Mouse Phenotyping Center, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Mi-Ock Lee
- Department of Pharmacy, College of Pharmacy and Bio-MAX Institute, Seoul National University 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| |
Collapse
|
20
|
Guidarelli A, Fiorani M, Cerioni L, Scotti M, Cantoni O. Arsenite induces DNA damage via mitochondrial ROS and induction of mitochondrial permeability transition. Biofactors 2017; 43:673-684. [PMID: 28703385 DOI: 10.1002/biof.1375] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 02/06/2023]
Abstract
Arsenite is an established DNA-damaging agent and human carcinogen. We initially selected conditions in which the metalloid causes DNA strand scission in the absence of detectable apoptotic DNA degradation in U937 cells. This response was suppressed by catalase and by treatments (rotenone and ascorbic acid), or manipulations (respiration-deficient phenotype), preventing the mitochondrial formation of O2-. ( mitoO2-.). MitoO2-., and its dismutation product, H2 O2 , are therefore critically involved in the arsenite-dependent DNA-damaging response. We then established a link between mitoO2-./H2 O2 and mitochondrial permeability transition (MPT), and found that this second event also promoted the formation of DNA-damaging species. As a consequence, the DNA damage induced by arsenite, in addition to being abolished by the aforementioned treatments/manipulations, was also significantly reduced by the MPT inhibitor cyclosporin A (CsA). A CsA-sensitive induction of p53 mRNA expression was also detected. Finally, evidence of CsA-sensitive DNA strand scission was also obtained in MCF-7, HT22, and NCTC-2544 cells. MitoO2-./H2 O2 therefore directly mediates DNA damage induced by arsenite and indirectly promotes the formation of additional DNA-damaging species via the induction of MPT. © 2017 BioFactors, 43(5):673-684, 2017.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Mara Fiorani
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Liana Cerioni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Maddalena Scotti
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Orazio Cantoni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| |
Collapse
|
21
|
O’Brien WG, Ling HS, Zhao Z, Lee CC. New insights on the regulation of the adenine nucleotide pool of erythrocytes in mouse models. PLoS One 2017; 12:e0180948. [PMID: 28746349 PMCID: PMC5528878 DOI: 10.1371/journal.pone.0180948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/13/2017] [Indexed: 11/18/2022] Open
Abstract
The observation that induced torpor in non-hibernating mammals could result from an increased AMP concentration in circulation led our investigation to reveal that the added AMP altered oxygen transport of erythrocytes. To further study the effect of AMP in regulation of erythrocyte function and systemic metabolism, we generated mouse models deficient in key erythrocyte enzymes in AMP metabolism. We have previously reported altered erythrocyte adenine nucleotide levels corresponding to altered oxygen saturation in mice deficient in both CD73 and AMPD3. Here we further investigate how these Ampd3-/-/Cd73-/- mice respond to the administered dose of AMP in comparison with the control models of single enzyme deficiency and wild type. We found that Ampd3-/-/Cd73-/- mice are more sensitive to AMP-induced hypometabolism than mice with a single enzyme deficiency, which are more sensitive than wild type. A dose-dependent rightward shift of erythrocyte p50 values in response to increasing amounts of extracellular AMP was observed. We provide further evidence for the direct uptake of AMP by erythrocytes that is insensitive to dipyridamole, a blocker for ENT1. The uptake of AMP by the erythrocytes remained linear at the highest concentration tested, 10mM. We also observed competitive inhibition of AMP uptake by ATP and ADP but not by the other nucleotides and metabolites tested. Importantly, our studies suggest that AMP uptake is associated with an erythrocyte ATP release that is partially sensitive to inhibition by TRO19622 and Ca++ ion. Taken together, our study suggests a novel mechanism by which erythrocytes recycle and maintain their adenine nucleotide pool through AMP uptake and ATP release.
Collapse
Affiliation(s)
- William G. O’Brien
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Han Shawn Ling
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Zhaoyang Zhao
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, United States of America
- * E-mail:
| | - Cheng Chi Lee
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, United States of America
| |
Collapse
|
22
|
Mutations in the Caenorhabditis elegans orthologs of human genes required for mitochondrial tRNA modification cause similar electron transport chain defects but different nuclear responses. PLoS Genet 2017; 13:e1006921. [PMID: 28732077 PMCID: PMC5544249 DOI: 10.1371/journal.pgen.1006921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 08/04/2017] [Accepted: 07/13/2017] [Indexed: 11/19/2022] Open
Abstract
Several oxidative phosphorylation (OXPHOS) diseases are caused by defects in the post-transcriptional modification of mitochondrial tRNAs (mt-tRNAs). Mutations in MTO1 or GTPBP3 impair the modification of the wobble uridine at position 5 of the pyrimidine ring and cause heart failure. Mutations in TRMU affect modification at position 2 and cause liver disease. Presently, the molecular basis of the diseases and why mutations in the different genes lead to such different clinical symptoms is poorly understood. Here we use Caenorhabditis elegans as a model organism to investigate how defects in the TRMU, GTPBP3 and MTO1 orthologues (designated as mttu-1, mtcu-1, and mtcu-2, respectively) exert their effects. We found that whereas the inactivation of each C. elegans gene is associated with a mild OXPHOS dysfunction, mutations in mtcu-1 or mtcu-2 cause changes in the expression of metabolic and mitochondrial stress response genes that are quite different from those caused by mttu-1 mutations. Our data suggest that retrograde signaling promotes defect-specific metabolic reprogramming, which is able to rescue the OXPHOS dysfunction in the single mutants by stimulating the oxidative tricarboxylic acid cycle flux through complex II. This adaptive response, however, appears to be associated with a biological cost since the single mutant worms exhibit thermosensitivity and decreased fertility and, in the case of mttu-1, longer reproductive cycle. Notably, mttu-1 worms also exhibit increased lifespan. We further show that mtcu-1; mttu-1 and mtcu-2; mttu-1 double mutants display severe growth defects and sterility. The animal models presented here support the idea that the pathological states in humans may initially develop not as a direct consequence of a bioenergetic defect, but from the cell’s maladaptive response to the hypomodification status of mt-tRNAs. Our work highlights the important association of the defect-specific metabolic rewiring with the pathological phenotype, which must be taken into consideration in exploring specific therapeutic interventions. Post-transcriptional modification of tRNAs is a universal process, thought to be essential for optimizing the functions of tRNAs. In humans, defects in the modification at position 2 (performed by protein TRMU) and 5 (carried out by proteins GTPBP3 and MTO1) of the uridine located at the wobble position of mitochondrial tRNAs (mt-tRNAs) cause oxidative phosphorylation (OXPHOS) dysfunction, and lead to liver and heart failure, respectively. However, the underlying mechanisms leading to pathogenesis are not well-known, and hence there is no molecular explanation for the different clinical phenotypes. We use Caenorhabditis elegans to compare in the same animal model and genetic background the effects of inactivating the TRMU, GTPBP3 and MTO1 orthologues on the phenotype and gene expression pattern of nuclear and mitochondrial DNA. Our data show that C. elegans responds to mt-tRNA hypomodification by changing in a defect-specific manner the expression of nuclear and mitochondrial genes, which leads, in all single mutants, to a rescue of the OXPHOS dysfunction that is associated with a biological cost. Our work suggests that pathology may develop as a consequence of the cell’s maladaptive response to the hypomodification status of mt-tRNAs.
Collapse
|
23
|
Victral DM, Dias HRA, Silva SQ, Baeta BEL, Aquino SF. Enhancement of anaerobic degradation of azo dye with riboflavin and nicotinamide adenine dinucleotide harvested by osmotic lysis of wasted fermentation yeasts. ENVIRONMENTAL TECHNOLOGY 2017; 38:483-494. [PMID: 27279330 DOI: 10.1080/09593330.2016.1198831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The study presented here aims at identifying the source of redox mediators (riboflavin), electron carriers nicotinamide adenine dinucleotide (NAD) and carbon to perform decolorization of azo dye under anaerobic conditions after osmotic shock pretreatment of residual yeast from industrial fermentation. Pretreatment conditions were optimized by Doehlert experiment, varying NaCl concentration, temperature, yeast density and time. After the optimization, the riboflavin concentration in the residual yeast lysate (RYL) was 46% higher than the one present in commercial yeast extract. Moreover, similar NAD concentration was observed in both extracts. Subsequently, two decolorization experiments were performed, that is, a batch experiment (48 h) and a kinetic experiment (102 h). The results of the batch experiment showed that the use of the RYL produced by the optimized method increased decolorization rates and led to color removal efficiencies similar to those found when using the commercial extract (∼80%) and from 23% to 50% higher when compared to the control (without redox mediators). Kinetics analysis showed that methane production was also higher in the presence of yeast extract and RYL, and biogas was mostly generated after stabilization of color removal. In all kinetics experiments the azo dye degradation followed the pseudo-second-order model, which suggested that there was a concomitant adsorption/degradation of the dye on the biomass cell surface. Therefore, results showed the possibility of applying the pretreated residual yeast to improve color removal under anaerobic conditions, which is a sustainable process.
Collapse
Affiliation(s)
- Davi M Victral
- a Postgraduation Program of Environmental Engineering (Proamb) , Federal University of Ouro Preto (UFOP) , Ouro Preto , Brazil
| | - Heitor R A Dias
- b Department of Environmental Engineering , Federal University of Ouro Preto (UFOP) , Ouro Preto , Brazil
| | - Silvana Q Silva
- c Department of Biological Sciences (DCBI) , Federal University of Ouro Preto (UFOP) , Ouro Preto , Brazil
| | - Bruno E L Baeta
- d Department of Chemistry (DEQUI) , Federal University of Ouro Preto (UFOP) , Ouro Preto , Brazil
| | - Sérgio F Aquino
- d Department of Chemistry (DEQUI) , Federal University of Ouro Preto (UFOP) , Ouro Preto , Brazil
| |
Collapse
|
24
|
Jimmerson LC, Bushman LR, Ray ML, Anderson PL, Kiser JJ. A LC-MS/MS Method for Quantifying Adenosine, Guanosine and Inosine Nucleotides in Human Cells. Pharm Res 2017; 34:73-83. [PMID: 27633886 PMCID: PMC5177511 DOI: 10.1007/s11095-016-2040-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/08/2016] [Indexed: 02/08/2023]
Abstract
PURPOSE To develop and validate a method for the simultaneous measurement of adenosine, guanosine, and inosine derived from mono (MP) and triphosphate (TP) forms in peripheral blood mononuclear cells (PBMCs), red blood cells (RBCs) and dried blood spots (DBS). METHODS Solid phase extraction of cell lysates followed by dephosphorylation to molar equivalent nucleoside and LC-MS/MS quantification. RESULTS The assay was linear for each of the three quantification ranges: 10-2000, 1.0-200 and 0.25-50 pmol/sample for adenosine, guanosine, and inosine, respectively. Intraassay (n = 6) and interassay (n = 18) precision (%CV) were within 1.7 to 16% while accuracy (%deviation) was within -11.5 to 14.7% for all three analytes. Nucleotide monophosphates were less concentrated than triphosphates (except for inosine) and levels in PBMCs were higher than RBCs for all three nucleotides (10, 55, and 5.6 fold for ATP, GTP and ITP, respectively). DBS samples had an average (SD) of -26% (22.6%) lower TP and 184% (173%) higher MP levels compared to paired RBC lysates, suggesting hydrolysis of the TP in DBS. CONCLUSION This method was accurate and precise for physiologically relevant concentrations of adenosine, guanosine and inosine nucleotides in mono- and triphosphate forms, providing a bioanalytical tool for quantitation of nucleotides for clinical studies.
Collapse
Affiliation(s)
- Leah C Jimmerson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 12850 E. Montview Blvd, V20 C238, Aurora, Colorado, 80045, USA
| | - Lane R Bushman
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 12850 E. Montview Blvd, V20 C238, Aurora, Colorado, 80045, USA
| | - Michelle L Ray
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 12850 E. Montview Blvd, V20 C238, Aurora, Colorado, 80045, USA
| | - Peter L Anderson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 12850 E. Montview Blvd, V20 C238, Aurora, Colorado, 80045, USA
| | - Jennifer J Kiser
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, 12850 E. Montview Blvd, V20 C238, Aurora, Colorado, 80045, USA.
| |
Collapse
|
25
|
Trifluoperazine inhibits acetaminophen-induced hepatotoxicity and hepatic reactive nitrogen formation in mice and in freshly isolated hepatocytes. Toxicol Rep 2017; 4:134-142. [PMID: 28503408 PMCID: PMC5426116 DOI: 10.1016/j.toxrep.2017.02.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Increased reactive nitrogen and oxygen species formation leads to APAP hepatoxicity. TFP is known to block nNOS both in vivo as well as in vitro. The nNOS inhibitor TFP blocks toxicity and the increased RNS/ROS formation. Toxicity occurs with increased 3- nitro tyrosine both in vivo as well as in vitro. NNOS inhibition by TFP leads to decreasing 3-nitro tyrosine in vivo as well as in vitro.
The hepatotoxicity of acetaminophen (APAP) occurs by initial metabolism to N-acetyl-p-benzoquinone imine which depletes GSH and forms APAP-protein adducts. Subsequently, the reactive nitrogen species peroxynitrite is formed from nitric oxide (NO) and superoxide leading to 3-nitrotyrosine in proteins. Toxicity occurs with inhibited mitochondrial function. We previously reported that in hepatocytes the nNOS (NOS1) inhibitor NANT inhibited APAP toxicity, reactive nitrogen and oxygen species formation, and mitochondrial dysfunction. In this work we examined the effect of trifluoperazine (TFP), a calmodulin antagonist that inhibits calcium induced nNOS activation, on APAP hepatotoxicity and reactive nitrogen formation in murine hepatocytes and in vivo. In freshly isolated hepatocytes TFP inhibited APAP induced toxicity, reactive nitrogen formation (NO, GSNO, and 3-nitrotyrosine in protein), reactive oxygen formation (superoxide), loss of mitochondrial membrane potential, decreased ATP production, decreased oxygen consumption rate, and increased NADH accumulation. TFP did not alter APAP induced GSH depletion in the hepatocytes or the formation of APAP protein adducts which indicated that reactive metabolite formation was not inhibited. Since we previously reported that TFP inhibits the hepatotoxicity of APAP in mice without altering hepatic APAP-protein adduct formation, we examined the APAP treated mouse livers for evidence of reactive nitrogen formation. 3-Nitrotyrosine in hepatic proteins and GSNO were significantly increased in APAP treated mouse livers and decreased in the livers of mice treated with APAP plus TFP. These data are consistent with a hypothesis that APAP hepatotoxicity occurs with altered calcium metabolism, activation of nNOS leading to increased reactive nitrogen formation, and mitochondrial dysfunction.
Collapse
|
26
|
Determination of nucleoside triphosphatase activities from measurement of true inorganic phosphate in the presence of labile phosphate compounds. Anal Biochem 2016; 520:62-67. [PMID: 28017740 DOI: 10.1016/j.ab.2016.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/10/2016] [Accepted: 12/14/2016] [Indexed: 11/24/2022]
Abstract
One of the most common assays for nucleoside triphosphatase (NTPase) activity entails the quantification of inorganic phosphate (Pi) as a colored phosphomolybdate complex at low pH. While this assay is very sensitive, it is not selective for Pi in the presence of labile organic phosphate compounds (OPCs). Since NTPase activity assays typically require a large excess of OPCs, such as nucleotides, selectivity for Pi in the presence of OPCs is often critical in evaluating enzyme activity. Here we present an improved method for the measurement of enzymatic nucleotide hydrolysis as Pi released, which achieves selectivity for Pi in the presence of OPCs while also avoiding the costs and hazards inherent in other methods for measuring nucleotide hydrolysis. We apply this method to the measurement of ATP hydrolysis by nitrogenase and GTP hydrolysis by elongation factor G (EF-G) in order to demonstrate the broad applicability of our method for the determination of nucleotide hydrolysis in the presence of interfering OPCs.
Collapse
|
27
|
Martins JD, Silva A, Ferreira I, Gonçalo M, Custódio JBA, Lopes MC, Domingues MRM, Neves BM, Cruz MT. Adenosine diphosphate involvement in THP-1 maturation triggered by the contact allergen 1-fluoro-2,4-dinitrobenzene. Toxicol Res (Camb) 2016; 5:1512-1521. [PMID: 30090452 PMCID: PMC6060794 DOI: 10.1039/c6tx00240d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/03/2016] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells' (DC) activation is considered a key event in the adverse outcome pathway for skin sensitization elicited by covalent binding of chemicals to proteins. The mechanisms underlying DC activation by contact sensitizers are not completely understood. However, several "danger signals" are pointed as relevant effectors. Among these extra-cellular early danger signals, purines may be crucial for the development of xenoinflammation and several reports indicate their involvement in contact allergic reactions. In the present work we used the DC-surrogate monocytic cell line THP-1, cultured alone or co-cultured with the human keratinocyte cell line HaCaT, to explore the contribution of extracellular adenine nucleotides to THP-1 maturation triggered by the extreme contact sensitizer, 1-fluoro-2,4-dinitrobenzene (DNFB). We found that THP-1 maturation induced by DNFB is impaired after purinergic signaling inhibition, and that the transcription of the purinergic metabotropic receptors P2Y2 and P2Y11 is modulated by the sensitizer. We also detected that THP-1 cells only partially hydrolyse extracellular adenosine triphosphate, leading to accumulation of the mono-phosphate derivative, AMP. We detected different and non-overlapping activation patterns of mitogen activated protein kinases by DNFB and extracellular nucleotides. Overall, our results indicate that THP-1 maturation induced by DNFB is strongly modulated by extracellular adenine nucleotides through metabotropic purinergic receptors. This knowledge unveils a molecular toxicity pathway evoked by sensitizers and involved in THP-1 maturation, a DC-surrogate cell line thoroughly used in in vitro tests for the identification of skin allergens.
Collapse
Affiliation(s)
- J D Martins
- Faculty of Pharmacy University of Coimbra , 3000-548 Coimbra , Portugal . ; ; ; Tel: +351 239 480 209
- CNC - Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - A Silva
- CNC - Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - I Ferreira
- CNC - Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - M Gonçalo
- Department of Dermatology , University Hospital and Faculty of Medicine , University of Coimbra , 3000-075 Coimbra , Portugal
| | - J B A Custódio
- Faculty of Pharmacy University of Coimbra , 3000-548 Coimbra , Portugal . ; ; ; Tel: +351 239 480 209
- CNC - Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - M C Lopes
- Faculty of Pharmacy University of Coimbra , 3000-548 Coimbra , Portugal . ; ; ; Tel: +351 239 480 209
- CNC - Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| | - M R M Domingues
- Department of Chemistry , Mass Spectrometry Center , QOPNA , University of Aveiro , Campus Universitário de Santiago , 3810-193 Aveiro , Portugal
| | - B M Neves
- Faculty of Pharmacy University of Coimbra , 3000-548 Coimbra , Portugal . ; ; ; Tel: +351 239 480 209
- CNC - Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
- Department of Chemistry , Mass Spectrometry Center , QOPNA , University of Aveiro , Campus Universitário de Santiago , 3810-193 Aveiro , Portugal
| | - M T Cruz
- Faculty of Pharmacy University of Coimbra , 3000-548 Coimbra , Portugal . ; ; ; Tel: +351 239 480 209
- CNC - Center for Neuroscience and Cell Biology , University of Coimbra , 3004-517 Coimbra , Portugal
| |
Collapse
|
28
|
Biagiotti S, Menotta M, Orazi S, Spapperi C, Brundu S, Fraternale A, Bianchi M, Rossi L, Chessa L, Magnani M. Dexamethasone improves redox state in ataxia telangiectasia cells by promoting an NRF2-mediated antioxidant response. FEBS J 2016; 283:3962-3978. [PMID: 27636396 DOI: 10.1111/febs.13901] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 09/12/2016] [Accepted: 09/14/2016] [Indexed: 12/17/2022]
Abstract
Ataxia telangiectasia (A-T) is a rare incurable neurodegenerative disease caused by biallelic mutations in the gene for ataxia-telangiectasia mutated (ATM). The lack of a functional ATM kinase leads to a pleiotropic phenotype, and oxidative stress is considered to have a crucial role in the complex physiopathology. Recently, steroids have been shown to reduce the neurological symptoms of the disease, although the molecular mechanism of this effect is largely unknown. In the present study, we have demonstrated that dexamethasone treatment of A-T lymphoblastoid cells increases the content of two of the most abundant antioxidants [glutathione (GSH) and NADPH] by up to 30%. Dexamethasone promoted the nuclear accumulation of the transcription factor nuclear factor (erythroid-derived 2)-like 2 to drive expression of antioxidant pathways involved in GSH synthesis and NADPH production. The latter effect was via glucose 6-phosphate dehydrogenase activation, as confirmed by increased enzyme activity and enhancement of the pentose phosphate pathway rate. This evidence indicates that glucocorticoids are able to potentiate antioxidant defenses to counteract oxidative stress in ataxia telangiectasia, and also reveals an unexpected role for dexamethasone in redox homeostasis and cellular antioxidant activity.
Collapse
Affiliation(s)
- Sara Biagiotti
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| | - Michele Menotta
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| | - Sara Orazi
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| | - Chiara Spapperi
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| | - Serena Brundu
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| | | | - Marzia Bianchi
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| | - Luigia Rossi
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| | - Luciana Chessa
- Department of Clinical and Molecular Medicine, University 'La Sapienza', Roma, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Italy
| |
Collapse
|
29
|
Banerjee S, Aykin-Burns N, Krager KJ, Shah SK, Melnyk SB, Hauer-Jensen M, Pawar SA. Loss of C/EBPδ enhances IR-induced cell death by promoting oxidative stress and mitochondrial dysfunction. Free Radic Biol Med 2016; 99:296-307. [PMID: 27554969 PMCID: PMC5673253 DOI: 10.1016/j.freeradbiomed.2016.08.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/26/2016] [Accepted: 08/17/2016] [Indexed: 01/01/2023]
Abstract
Exposure of cells to ionizing radiation (IR) generates reactive oxygen species (ROS). This results in increased oxidative stress and DNA double strand breaks (DSBs) which are the two underlying mechanisms by which IR causes cell/tissue injury. Cells that are deficient or impaired in the cellular antioxidant response are susceptible to IR-induced apoptosis. The transcription factor CCAAT enhancer binding protein delta (Cebpd, C/EBPδ) has been implicated in the regulation of oxidative stress, DNA damage response, genomic stability and inflammation. We previously reported that Cebpd-deficient mice are sensitive to IR and display intestinal and hematopoietic injury, however the underlying mechanism is not known. In this study, we investigated whether an impaired ability to detoxify IR-induced ROS was the underlying cause of the increased radiosensitivity of Cebpd-deficient cells. We found that Cebpd-knockout (KO) mouse embryonic fibroblasts (MEFs) expressed elevated levels of ROS, both at basal levels and after exposure to gamma radiation which correlated with increased apoptosis, and decreased clonogenic survival. Pre-treatment of wild type (WT) and KO MEFs with polyethylene glycol-conjugated Cu-Zn superoxide dismutase (PEG-SOD) and catalase (PEG-CAT) combination prior to irradiation showed a partial rescue of clonogenic survival, thus demonstrating a role for increased intracellular oxidants in promoting IR-induced cell death. Analysis of mitochondrial bioenergetics revealed that irradiated KO MEFs showed significant reductions in basal, adenosine triphosphate (ATP)-linked, maximal respiration and reserved respiratory capacity and decrease in intracellular ATP levels compared to WT MEFs indicating they display mitochondrial dysfunction. KO MEFs expressed significantly lower levels of the cellular antioxidant glutathione (GSH) and its precursor- cysteine as well as methionine. In addition to its antioxidant function, GSH plays an important role in detoxification of lipid peroxidation products such as 4-hydroxynonenal (4-HNE). The reduced GSH levels observed in KO MEFs correlated with elevated levels of 4-HNE protein adducts in irradiated KO MEFs compared to respective WT MEFs. We further showed that pre-treatment with the GSH precursor, N-acetyl L-cysteine (NAC) prior to irradiation showed a significant reduction of IR-induced cell death and increases in GSH levels, which contributed to the overall increase in clonogenic survival of KO MEFs. In contrast, pre-treatment with the GSH synthesis inhibitor- buthionine sulfoximine (BSO) further reduced the clonogenic survival of irradiated KO MEFs. This study demonstrates a novel role for C/EBPδ in protection from basal as well as IR-induced oxidative stress and mitochondrial dysfunction thus promoting post-radiation survival.
Collapse
Affiliation(s)
- Sudip Banerjee
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Kimberly J Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Sumit K Shah
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Stepan B Melnyk
- Arkansas Children's Hospital Research Institute, Little Rock, AR 72205, United States
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States; Surgical Services, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, United States
| | - Snehalata A Pawar
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
30
|
Naia L, Ribeiro M, Rodrigues J, Duarte AI, Lopes C, Rosenstock TR, Hayden MR, Rego AC. Insulin and IGF-1 regularize energy metabolites in neural cells expressing full-length mutant huntingtin. Neuropeptides 2016; 58:73-81. [PMID: 26876526 DOI: 10.1016/j.npep.2016.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/07/2016] [Accepted: 01/31/2016] [Indexed: 01/08/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder linked to the expression of mutant huntingtin. Bioenergetic dysfunction has been described to contribute to HD pathogenesis. Thus, treatment paradigms aimed to ameliorate energy deficits appear to be suitable candidates in HD. In previous studies, we observed protective effects of insulin growth factor-1 (IGF-1) in YAC128 and R6/2 mice, two HD mouse models, whereas IGF-1 and/or insulin halted mitochondrial-driven oxidative stress in mutant striatal cells and mitochondrial dysfunction in HD human lymphoblasts. Here, we analyzed the effect of IGF-1 versus insulin on energy metabolic parameters using striatal cells derived from HD knock-in mice and primary cortical cultures from YAC128 mice. STHdh(Q111/Q111) cells exhibited decreased ATP/ADP ratio and increased phosphocreatine levels. Moreover, pyruvate levels were increased in mutant cells, most probably in consequence of a decrease in pyruvate dehydrogenase (PDH) protein expression and increased PDH phosphorylation, reflecting its inactivation. Insulin and IGF-1 treatment significantly decreased phosphocreatine levels, whereas IGF-1 only decreased pyruvate levels in mutant cells. In a different scenario, primary cortical cultures derived from YAC128 mice also displayed energetic abnormalities. We observed a decrease in both ATP/ADP and phosphocreatine levels, which were prevented following exposure to insulin or IGF-1. Furthermore, decreased lactate levels in YAC128 cultures occurred concomitantly with a decline in lactate dehydrogenase activity, which was ameliorated with both insulin and IGF-1. These data demonstrate differential HD-associated metabolic dysfunction in striatal cell lines and primary cortical cultures, both of which being alleviated by insulin and IGF-1.
Collapse
Affiliation(s)
- Luana Naia
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal
| | - Márcio Ribeiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Joana Rodrigues
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Ana I Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Polo II, Coimbra, Portugal
| | - Carla Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Polo II, Coimbra, Portugal
| | - Tatiana R Rosenstock
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Polo II, Coimbra, Portugal
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - A Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal.
| |
Collapse
|
31
|
Alexandrino H, Varela AT, Teodoro JS, Martins MA, Rolo AP, Tralhão JG, Palmeira CM, Castro E Sousa F. Mitochondrial bioenergetics and posthepatectomy liver dysfunction. Eur J Clin Invest 2016; 46:627-35. [PMID: 27138992 DOI: 10.1111/eci.12639] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 04/29/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Liver regeneration requires an enormous energy supply. Experimental evidence suggests that mitochondrial function is of paramount importance for liver regeneration. However, this has not been investigated in the clinical setting. We aimed to: (i) evaluate changes in mitochondrial function during hepatectomy, especially after hepatic pedicle clamping; and (ii) correlate these changes with postoperative hepatocellular function and clinical outcome. MATERIALS AND METHODS Prospective study of thirty patients undergoing hepatectomy. Measurement of mitochondrial membrane potential, respiration and adenosine triphosphate content in intra-operative liver biopsies performed in nonresected parenchyma. Correlation of findings with duration of hepatic pedicle clamping, postoperative markers of hepatocellular necrosis and function (aminotransferases, arterial lactate, international normalized ratio, bilirubin), and morbidity. RESULTS Longer hepatic pedicle clamping was associated with worse mitochondrial depolarization (r = -0·519; P = 0·011) and longer lag phase (r = 0·568; P = 0·006). Higher postoperative peak aminotransferases, international normalized ratio and bilirubin correlated with worse mitochondrial function (P < 0·05). After major hepatectomy, mitochondrial respiration correlated with postoperative arterial lactate clearance (r = 0·756; P = 0·049). Mitochondrial bioenergetic parameters were significantly decreased in patients with liver-specific morbidity and postoperative liver failure (P < 0·05). On multivariate analysis, decrease in mitochondrial potential was an independent risk factor for liver-specific morbidity (OR = 13·7; P = 0·043). Worse lag phase was highly predictive of posthepatectomy liver failure (area under the curve: 0·933; P = 0·008). CONCLUSIONS There is a relationship between mitochondrial function, duration of hepatic pedicle clamping and clinical outcome after hepatectomy. Mitochondrial bioenergetics can potentially translate into clinical practice, assisting in earlier diagnosis of postoperative liver dysfunction, and as a target for future pharmacological therapies.
Collapse
Affiliation(s)
- Henrique Alexandrino
- Department of Surgery A, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Clínica Universitária de Cirurgia III, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana T Varela
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - João Soeiro Teodoro
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Mónica A Martins
- Department of Surgery A, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Clínica Universitária de Cirurgia III, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Anabela P Rolo
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Guilherme Tralhão
- Department of Surgery A, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Clínica Universitária de Cirurgia III, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Investigation on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Center of Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Francisco Castro E Sousa
- Department of Surgery A, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Clínica Universitária de Cirurgia III, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Investigation on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
32
|
Manoj KM, Parashar A, Gade SK, Venkatachalam A. Functioning of Microsomal Cytochrome P450s: Murburn Concept Explains the Metabolism of Xenobiotics in Hepatocytes. Front Pharmacol 2016; 7:161. [PMID: 27445805 PMCID: PMC4918403 DOI: 10.3389/fphar.2016.00161] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 05/31/2016] [Indexed: 11/13/2022] Open
Abstract
Using oxygen and NADPH, the redox enzymes cytochrome P450 (CYP) and its reductase (CPR) work in tandem to carry out the phase I metabolism of a vast majority of drugs and xenobiotics. As per the erstwhile understanding of the catalytic cycle, binding of the substrate to CYP's heme distal pocket allows CPR to pump electrons through a CPR-CYP complex. In turn, this trigger (a thermodynamic push of electrons) leads to the activation of oxygen at CYP's heme-center, to give Compound I, a two-electron deficient enzyme reactive intermediate. The formation of diffusible radicals and reactive oxygen species (DROS, hitherto considered an undesired facet of the system) was attributed to the heme-center. Recently, we had challenged these perceptions and proposed the murburn ("mured burning" or "mild unrestricted burning") concept to explain heme enzymes' catalytic mechanism, electron-transfer phenomena and the regulation of redox equivalents' consumption. Murburn concept incorporates a one-electron paradigm, advocating obligatory roles for DROS. The new understanding does not call for high-affinity substrate-binding at the heme distal pocket of the CYP (the first and the most crucial step of the erstwhile paradigm) or CYP-CPR protein-protein complexations (the operational backbone of the erstwhile cycle). Herein, the dynamics of reduced nicotinamide nucleotides' consumption, peroxide formation and depletion, product(s) formation, etc. was investigated with various controls, by altering reaction variables, environments and through the incorporation of diverse molecular probes. In several CYP systems, control reactions lacking the specific substrate showed comparable or higher peroxide in milieu, thereby discrediting the foundations of the erstwhile hypothesis. The profiles obtained by altering CYP:CPR ratios and the profound inhibitions observed upon the incorporation of catalytic amounts of horseradish peroxidase confirm the obligatory roles of DROS in milieu, ratifying murburn as the operative concept. The mechanism of uncoupling (peroxide/water formation) was found to be dependent on multiple one and two electron equilibriums amongst the reaction components. The investigation explains the evolutionary implications of xenobiotic metabolism, confirms the obligatory role of diffusible reactive species in routine redox metabolism within liver microsomes and establishes that a redox enzyme like CYP enhances reaction rates (achieves catalysis) via a novel (hitherto unknown) modality.
Collapse
Affiliation(s)
| | - Abhinav Parashar
- Hemoproteins Lab, School of Bio Sciences and Technology, VIT University Vellore, India
| | - Sudeep K Gade
- Hemoproteins Lab, School of Bio Sciences and Technology, VIT University Vellore, India
| | | |
Collapse
|
33
|
Cai H, Scott E, Kholghi A, Andreadi C, Rufini A, Karmokar A, Britton RG, Horner-Glister E, Greaves P, Jawad D, James M, Howells L, Ognibene T, Malfatti M, Goldring C, Kitteringham N, Walsh J, Viskaduraki M, West K, Miller A, Hemingway D, Steward WP, Gescher AJ, Brown K. Cancer chemoprevention: Evidence of a nonlinear dose response for the protective effects of resveratrol in humans and mice. Sci Transl Med 2016. [PMID: 26223300 DOI: 10.1126/scitranslmed.aaa7619] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Resveratrol is widely promoted as a potential cancer chemopreventive agent, but a lack of information on the optimal dose prohibits rationally designed trials to assess efficacy. To challenge the assumption that "more is better," we compared the pharmacokinetics and activity of a dietary dose with an intake 200 times higher. The dose-response relationship for concentrations generated and the metabolite profile of [(14)C]-resveratrol in colorectal tissue of cancer patients helped us to define clinically achievable levels. In Apc(Min) mice (a model of colorectal carcinogenesis) that received a high-fat diet, the low resveratrol dose suppressed intestinal adenoma development more potently than did the higher dose. Efficacy correlated with activation of adenosine monophosphate-activated protein kinase (AMPK) and increased expression of the senescence marker p21. Nonlinear dose responses were observed for AMPK and mechanistic target of rapamycin (mTOR) signaling in mouse adenoma cells, culminating in autophagy and senescence. In human colorectal tissues exposed to low dietary concentrations of resveratrol ex vivo, we measured enhanced AMPK phosphorylation and autophagy. The expression of the cytoprotective NAD(P)H dehydrogenase, quinone 1 (NQO1) enzyme was also increased in tissues from cancer patients participating in our [(14)C]-resveratrol trial. These findings warrant a revision of developmental strategies for diet-derived agents designed to achieve cancer chemoprevention.
Collapse
Affiliation(s)
- Hong Cai
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Edwina Scott
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Abeer Kholghi
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Catherine Andreadi
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Alessandro Rufini
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Ankur Karmokar
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Robert G Britton
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Emma Horner-Glister
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Peter Greaves
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Dhafer Jawad
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Mark James
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Lynne Howells
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Ted Ognibene
- Lawrence Livermore National Laboratory, 7000 East Avenue, Livermore, CA 94551, USA
| | - Michael Malfatti
- Lawrence Livermore National Laboratory, 7000 East Avenue, Livermore, CA 94551, USA
| | - Christopher Goldring
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, UK
| | - Neil Kitteringham
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, UK
| | - Joanne Walsh
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, UK
| | - Maria Viskaduraki
- Bioinformatics and Biostatistics Support Hub, University of Leicester, Maurice Shock Building, Leicester LE1 9HN, UK
| | - Kevin West
- University Hospitals of Leicester NHS Trust, Leicester LE1 5WW, UK
| | - Andrew Miller
- University Hospitals of Leicester NHS Trust, Leicester LE1 5WW, UK
| | - David Hemingway
- University Hospitals of Leicester NHS Trust, Leicester LE1 5WW, UK
| | - William P Steward
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Andreas J Gescher
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK
| | - Karen Brown
- Cancer Chemoprevention Group, Department of Cancer Studies, University of Leicester, Leicester LE2 7LX, UK.
| |
Collapse
|
34
|
Zhu Z, Lu Q, Zeng F, Wang J, Huang S. Compatibility between mitochondrial and nuclear genomes correlates with the quantitative trait of lifespan in Caenorhabditis elegans. Sci Rep 2015; 5:17303. [PMID: 26601686 PMCID: PMC4658563 DOI: 10.1038/srep17303] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/28/2015] [Indexed: 12/12/2022] Open
Abstract
Mutations in mitochondrial genome have epistatic effects on organisms depending on
the nuclear background, but a role for the compatibility of mitochondrial-nuclear
genomes (mit-n) in the quantitative nature of a complex trait remains unexplored. We
studied a panel of recombinant inbred advanced intercrossed lines (RIAILs) of C.
elegans that were established from a cross between the N2 and HW strains. We
determined the HW nuclear genome content and the mitochondrial type (HW or N2) of
each RIAIL strain. We found that the degree of mit-n compatibility was correlated
with the lifespans but not the foraging behaviors of RIAILs. Several known
aging-associated QTLs individually showed no relationship with mitotypes but
collectively a weak trend consistent with a role in mit-n compatibility. By
association mapping, we identified 293 SNPs that showed linkage with lifespan and a
relationship with mitotypes consistent with a role in mit-n compatibility. We
further found an association between mit-n compatibility and several functional
characteristics of mitochondria as well as the expressions of genes involved in the
respiratory oxidation pathway. The results provide the first evidence implicating
mit-n compatibility in the quantitative nature of a complex trait, and may be
informative to certain evolutionary puzzles on hybrids.
Collapse
Affiliation(s)
- Zuobin Zhu
- State Key Laboratory of Medical Genetics, School of Life Sciences, Xiangya Medical School, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078, China
| | - Qing Lu
- State Key Laboratory of Medical Genetics, School of Life Sciences, Xiangya Medical School, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078, China
| | - Fangfang Zeng
- State Key Laboratory of Medical Genetics, School of Life Sciences, Xiangya Medical School, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078, China
| | - Junjing Wang
- State Key Laboratory of Medical Genetics, School of Life Sciences, Xiangya Medical School, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078, China
| | - Shi Huang
- State Key Laboratory of Medical Genetics, School of Life Sciences, Xiangya Medical School, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078, China
| |
Collapse
|
35
|
Testosterone deficiency induced by progressive stages of diabetes mellitus impairs glucose metabolism and favors glycogenesis in mature rat Sertoli cells. Int J Biochem Cell Biol 2015; 66:1-10. [DOI: 10.1016/j.biocel.2015.07.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 06/10/2015] [Accepted: 07/01/2015] [Indexed: 12/20/2022]
|
36
|
Rodrigues AS, Pereira SL, Correia M, Gomes A, Perestrelo T, Ramalho-Santos J. Differentiate or Die: 3-Bromopyruvate and Pluripotency in Mouse Embryonic Stem Cells. PLoS One 2015; 10:e0135617. [PMID: 26266544 PMCID: PMC4534445 DOI: 10.1371/journal.pone.0135617] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/24/2015] [Indexed: 02/06/2023] Open
Abstract
Background Pluripotent embryonic stem cells grown under standard conditions (ESC) have a markedly glycolytic profile, which is shared with many different types of cancer cells. Thus, some therapeutic strategies suggest that pharmacologically shifting cancer cells towards an oxidative phenotype, using glycolysis inhibitors, may reduce cancer aggressiveness. Given the metabolic parallels between cancer and stemness would chemotherapeutical agents have an effect on pluripotency, and could a strategy involving these agents be envisioned to modulate stem cell fate in an accessible manner? In this manuscript we attempted to determine the effects of 3-bromopyruvate (3BrP) in pluripotency. Although it has other intracellular targets, this compound is a potent inhibitor of glycolysis enzymes thought to be important to maintain a glycolytic profile. The goal was also to determine if we could contribute towards a pharmacologically accessible metabolic strategy to influence cell differentiation. Methodology/Principal Findings Mouse embryonic stem cells (mESC) grown under standard pluripotency conditions (in the presence of Leukemia Inducing Factor- LIF) were treated with 3BrP. As a positive control for differentiation other mESCs were grown without LIF. Overall our results demonstrate that 3BrP negatively affects pluripotency, forcing cells to become less glycolytic and with more active mitochondria. These changes in metabolism are correlated with increased differentiation, even under pluripotency conditions (i.e. in the presence of LIF). However, 3BrP also significantly impaired cell function, and may have other roles besides affecting the metabolic profile of mESCs. Conclusions/Findings Treatment of mESCs with 3BrP triggered a metabolic switch and loss of pluripotency, even in the presence of LIF. Interestingly, the positive control for differentiation allowed for a distinction between 3BrP effects and changes associated with spontaneous differentiation/loss of pluripotency in the absence of LIF. Additionally, there was a slight differentiation bias towards mesoderm in the presence of 3BrP. However, the side effects on cellular function suggest that the use of this drug is probably not adequate to efficiently push cells towards specific differentiation fates.
Collapse
Affiliation(s)
- Ana Sofia Rodrigues
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Sandro L. Pereira
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Marcelo Correia
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Andreia Gomes
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
- Biocant—Center of Innovation in Biotechnology, Cantanhede, Portugal
| | - Tânia Perestrelo
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - João Ramalho-Santos
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- * E-mail:
| |
Collapse
|
37
|
O'Brien WG, Berka V, Tsai AL, Zhao Z, Lee CC. CD73 and AMPD3 deficiency enhance metabolic performance via erythrocyte ATP that decreases hemoglobin oxygen affinity. Sci Rep 2015; 5:13147. [PMID: 26249166 PMCID: PMC4650700 DOI: 10.1038/srep13147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/22/2015] [Indexed: 12/15/2022] Open
Abstract
Erythrocytes are the key target in 5′-AMP induced hypometabolism. To understand how regulation of endogenous erythrocyte AMP levels modulates systemic metabolism, we generated mice deficient in both CD73 and AMPD3, the key catabolic enzymes for extracellular and intra-erythrocyte AMP, respectively. Under physiological conditions, these mice displayed enhanced capacity for physical activity accompanied by significantly higher food and oxygen consumption, compared to wild type mice. Erythrocytes from Ampd3−/− mice exhibited higher half-saturation pressure of oxygen (p50) and about 3-fold higher levels of ATP and ADP, while they maintained normal 2,3-bisphosphoglycerate (2,3-BPG), methemoglobin levels and intracellular pH. The affinity of mammalian hemoglobin for oxygen is thought to be regulated primarily by 2,3-BPG levels and pH (the Bohr effect). However, our results show that increased endogenous levels of ATP and ADP, but not AMP, directly increase the p50 value of hemoglobin. Additionally, the rise in erythrocyte p50 directly correlates with an enhanced capability of systemic metabolism.
Collapse
Affiliation(s)
- William G O'Brien
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, Texas, USA 77030
| | - Vladimir Berka
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas, USA 77030
| | - Ah-Lim Tsai
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center, Houston, Texas, USA 77030
| | - Zhaoyang Zhao
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, Texas, USA 77030
| | - Cheng Chi Lee
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, Texas, USA 77030
| |
Collapse
|
38
|
Rodrigues AS, Correia M, Gomes A, Pereira SL, Perestrelo T, Sousa MI, Ramalho-Santos J. Dichloroacetate, the Pyruvate Dehydrogenase Complex and the Modulation of mESC Pluripotency. PLoS One 2015; 10:e0131663. [PMID: 26147621 PMCID: PMC4493017 DOI: 10.1371/journal.pone.0131663] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/05/2015] [Indexed: 02/06/2023] Open
Abstract
Introduction The pyruvate dehydrogenase (PDH) complex is localized in the mitochondrial matrix catalyzing the irreversible decarboxylation of pyruvate to acetyl-CoA and NADH. For proper complex regulation the E1-α subunit functions as an on/off switch regulated by phosphorylation/dephosphorylation. In different cell types one of the four-pyruvate dehydrogenase kinase isoforms (PDHK1-4) can phosphorylate this subunit leading to PDH inactivation. Our previous results with human Embryonic Stem Cells (hESC), suggested that PDHK could be a key regulator in the metabolic profile of pluripotent cells, as it is upregulated in pluripotent stem cells. Therefore, we wondered if metabolic modulation, via inexpensive pharmacological inhibition of PDHK, could impact metabolism and pluripotency. Methods/Results In order to assess the importance of the PDH cycle in mouse Embryonic Stem Cells (mESC), we incubated cells with the PDHK inhibitor dichloroacetate (DCA) and observed that in its presence ESC started to differentiate. Changes in mitochondrial function and proliferation potential were also found and protein levels for PDH (both phosphorylated and non-phosphorylated) and PDHK1 were monitored. Interestingly, we were also able to describe a possible pathway that involves Hif-1α and p53 during DCA-induced loss of pluripotency. Results with ESCs treated with DCA were comparable to those obtained for cells grown without Leukemia Inhibitor Factor (LIF), used in this case as a positive control for differentiation. Conclusions DCA negatively affects ESC pluripotency by changing cell metabolism and elements related to the PDH cycle, suggesting that PDHK could function as a possible metabolic gatekeeper in ESC, and may be a good target to modulate metabolism and differentiation. Although further molecular biology-based experiments are required, our data suggests that inactive PDH favors pluripotency and that ESC have similar strategies as cancer cells to maintain a glycolytic profile, by using some of the signaling pathways found in the latter cells.
Collapse
Affiliation(s)
- Ana Sofia Rodrigues
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Marcelo Correia
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Andreia Gomes
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
- Biocant—Center of Innovation in Biotechnology, Cantanhede, Portugal
| | - Sandro L. Pereira
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Tânia Perestrelo
- PhD Programme in Experimental Biology and Biomedicine, CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Maria Inês Sousa
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - João Ramalho-Santos
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- * E-mail:
| |
Collapse
|
39
|
Aragon-Martinez OH, Galicia O, Isiordia-Espinoza MA, Martinez-Morales F. A novel method for measuring the ATP-related compounds in human erythrocytes. TOHOKU J EXP MED 2015; 233:205-14. [PMID: 25048613 DOI: 10.1620/tjem.233.205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The ATP-related compounds in whole blood or red blood cells have been used to evaluate the energy status of erythrocytes and the degradation level of the phosphorylated compounds under various conditions, such as chronic renal failure, drug monitoring, cancer, exposure to environmental toxics, and organ preservation. The complete interpretation of the energetic homeostasis of erythrocytes is only performed using the compounds involved in the degradation pathway for adenine nucleotides alongside the uric acid value. For the first time, we report a liquid chromatographic method using a diode array detector that measures all of these compounds in a small human whole blood sample (125 μL) within an acceptable time of 20 min. The stability was evaluated for all of the compounds and ranged from 96.3 to 105.1% versus the day zero values. The measurement had an adequate sensitivity for the ATP-related compounds (detection limits from 0.001 to 0.097 μmol/L and quantification limits from 0.004 to 0.294 μmol/L). This method is particularly useful for measuring inosine monophosphate, inosine, hypoxanthine, and uric acid. Moreover, this assay had acceptable linearity (r > 0.990), precision (coefficients of variation ranged from 0.1 to 2.0%), specificity (similar retention times and spectra in all samples) and recoveries (ranged from 89.2 to 104.9%). The newly developed method is invaluable for assessing the energetic homeostasis of red blood cells under diverse conditions, such as in vitro experiments and clinical settings.
Collapse
|
40
|
Effect of lysine acetylsalicylate on aluminium accumulation and (Na+/K+)ATPase activity in rat brain cortex synaptosomes after aluminium ingestion. Toxicol Lett 2015; 232:167-74. [DOI: 10.1016/j.toxlet.2014.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 10/09/2014] [Accepted: 10/09/2014] [Indexed: 11/21/2022]
|
41
|
Adenosine amine congener as a cochlear rescue agent. BIOMED RESEARCH INTERNATIONAL 2014; 2014:841489. [PMID: 25243188 PMCID: PMC4160640 DOI: 10.1155/2014/841489] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/29/2014] [Indexed: 12/20/2022]
Abstract
We have previously shown that adenosine amine congener (ADAC), a selective A1 adenosine receptor agonist, can ameliorate noise- and cisplatin-induced cochlear injury. Here we demonstrate the dose-dependent rescue effects of ADAC on noise-induced cochlear injury in a rat model and establish the time window for treatment. Methods. ADAC (25–300 μg/kg) was administered intraperitoneally to Wistar rats (8–10 weeks old) at intervals (6–72 hours) after exposure to traumatic noise (8–16 kHz, 110 dB sound pressure level, 2 hours). Hearing sensitivity was assessed using auditory brainstem responses (ABR) before and 12 days after noise exposure. Pharmacokinetic studies investigated ADAC concentrations in plasma after systemic (intravenous) administration. Results. ADAC was most effective in the first 24 hours after noise exposure at doses >50 μg/kg, providing up to 21 dB protection (averaged across 8–28 kHz). Pharmacokinetic studies demonstrated a short (5 min) half-life of ADAC in plasma after intravenous administration without detection of degradation products. Conclusion. Our data show that ADAC mitigates noise-induced hearing loss in a dose- and time-dependent manner, but further studies are required to establish its translation as a clinical otological treatment.
Collapse
|
42
|
Activation of IGF-1 and Insulin Signaling Pathways Ameliorate Mitochondrial Function and Energy Metabolism in Huntington’s Disease Human Lymphoblasts. Mol Neurobiol 2014; 51:331-48. [DOI: 10.1007/s12035-014-8735-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/29/2014] [Indexed: 12/31/2022]
|
43
|
Lu L, Qian Y, Wang L, Ma K, Zhang Y. Metal-enhanced fluorescence-based core-shell Ag@SiO₂ nanoflares for affinity biosensing via target-induced structure switching of aptamer. ACS APPLIED MATERIALS & INTERFACES 2014; 6:1944-50. [PMID: 24480015 DOI: 10.1021/am4049942] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
One of the great challenges in metal-enhanced fluorescence (MEF) technology is the achievement of distance modulation with nanometer accuracy between the fluorophore and metal surface to obtain maximum enhancement. We propose an MEF-based core-shell Ag@SiO2 nanoflare for distance control via the thickness of silica shell with cooperation of DNA hybridization. The nanoflare contains a 50 nm spherical silver nanoparticle (Ag NP) core, a 8 nm silica shell, and cyanine (Cy5)-labeled aptamer hybridized with a complementary DNA (cDNA) immobilized onto the shell surface. The formation of the Cy5-labeled aptamer/cDNA duplex on the Ag@SiO2 NP surface results in the confinement of Cy5 to the shell surface and an increase in the fluorescence of Cy5 with a 32-fold enhancement factor in bulk solution (signal-on). In the presence of affinity-binding targets, the Cy5-labeled aptamers confined onto the Ag@SiO2 NP surface dissociate from their cDNA into the solution because of structure switching. The target-induced release of aptamer leads to a reduction in the enhanced fluorescence signal of the labeled Cy5 moiety (signal-off). Thus, the nanoflare can be used as a sensor for target recognition. Using adenosine-5'-triphosphate (ATP) aptamer, detection of ATP has a linear response from 0 to 0.5 mM and a detection limit of 8 μM. With various types of DNA probes immobilized onto the core-shell Ag@SiO2 NPs, the MEF-based nanoflare has provided an effective platform for the detection and quantification of a broad range of analytes, such as mRNA regulation and detection, cell sorting, and gene profiling.
Collapse
Affiliation(s)
- Lu Lu
- Key Laboratory of Applied Surface and Colloid Chemistry of Ministry of Education and ‡Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University , Xi'an 710062, China
| | | | | | | | | |
Collapse
|
44
|
Rose S, Frye RE, Slattery J, Wynne R, Tippett M, Pavliv O, Melnyk S, James SJ. Oxidative stress induces mitochondrial dysfunction in a subset of autism lymphoblastoid cell lines in a well-matched case control cohort. PLoS One 2014; 9:e85436. [PMID: 24416410 PMCID: PMC3885720 DOI: 10.1371/journal.pone.0085436] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 11/26/2013] [Indexed: 01/26/2023] Open
Abstract
There is increasing recognition that mitochondrial dysfunction is associated with the autism spectrum disorders. However, little attention has been given to the etiology of mitochondrial dysfunction or how mitochondrial abnormalities might interact with other physiological disturbances associated with autism, such as oxidative stress. In the current study we used respirometry to examine reserve capacity, a measure of the mitochondrial ability to respond to physiological stress, in lymphoblastoid cell lines (LCLs) derived from children with autistic disorder (AD) as well as age and gender-matched control LCLs. We demonstrate, for the first time, that LCLs derived from children with AD have an abnormal mitochondrial reserve capacity before and after exposure to increasingly higher concentrations of 2,3-dimethoxy-1,4-napthoquinone (DMNQ), an agent that increases intracellular reactive oxygen species (ROS). Specifically, the AD LCLs exhibit a higher reserve capacity at baseline and a sharper depletion of reserve capacity when ROS exposure is increased, as compared to control LCLs. Detailed investigation indicated that reserve capacity abnormalities seen in AD LCLs were the result of higher ATP-linked respiration and maximal respiratory capacity at baseline combined with a marked increase in proton leak respiration as ROS was increased. We further demonstrate that these reserve capacity abnormalities are driven by a subgroup of eight (32%) of 25 AD LCLs. Additional investigation of this subgroup of AD LCLs with reserve capacity abnormalities revealed that it demonstrated a greater reliance on glycolysis and on uncoupling protein 2 to regulate oxidative stress at the inner mitochondria membrane. This study suggests that a significant subgroup of AD children may have alterations in mitochondrial function which could render them more vulnerable to a pro-oxidant microenvironment derived from intrinsic and extrinsic sources of ROS such as immune activation and pro-oxidant environmental toxicants. These findings are consistent with the notion that AD is caused by a combination of genetic and environmental factors.
Collapse
Affiliation(s)
- Shannon Rose
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
| | - Richard E. Frye
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
- * E-mail:
| | - John Slattery
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
| | - Rebecca Wynne
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
| | - Marie Tippett
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
| | - Oleksandra Pavliv
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
| | - Stepan Melnyk
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
| | - S. Jill James
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas, United States of America
| |
Collapse
|
45
|
Leoni L, Landini P. Microbiological methods for target-oriented screening of biofilm inhibitors. Methods Mol Biol 2014; 1147:175-186. [PMID: 24664833 DOI: 10.1007/978-1-4939-0467-9_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The ability of many pathogenic bacteria to grow as a biofilm results in lower susceptibility to antibiotic treatments and to the host immune response, thus leading to the development of chronic infections. The understanding that biofilms can play an important role in bacterial virulence has prompted the search for inhibitors of biofilm development and of biofilm-related cellular processes. In this report, we present two examples of target-based microbiological screenings for antimicrobials endowed with anti-biofilm activity, aimed respectively at the inhibition of the signal molecule cyclic di-GMP and of quorum sensing.
Collapse
Affiliation(s)
- Livia Leoni
- Department of Sciences, Università "Roma Tre", Rome, Italy
| | | |
Collapse
|
46
|
Rato L, Duarte AI, Tomás GD, Santos MS, Moreira PI, Socorro S, Cavaco JE, Alves MG, Oliveira PF. Pre-diabetes alters testicular PGC1-α/SIRT3 axis modulating mitochondrial bioenergetics and oxidative stress. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1837:335-44. [PMID: 24361842 DOI: 10.1016/j.bbabio.2013.12.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/10/2013] [Accepted: 12/12/2013] [Indexed: 11/19/2022]
Abstract
Pre-diabetes, a risk factor for type 2 diabetes development, leads to metabolic changes at testicular level. Peroxisome proliferator-activated receptor γ coactivator 1 α (PGC-1α) and Sirtuin 3 (Sirt3) are pivotal in mitochondrial function. We hypothesized that pre-diabetes disrupts testicular PGC-1α/Sirt3 axis, compromising testicular mitochondrial function. Using a high-energy-diet induced pre-diabetic rat model, we evaluated testicular levels of PGC-1α and its downstream targets, nuclear respiratory factors 1 (NRF-1) and 2 (NRF-2), mitochondrial transcription factor A (TFAM) and Sirt3. We also assessed mitochondrial DNA (mtDNA) content, mitochondrial function, energy levels and oxidative stress parameters. Protein levels were quantified by Western Blot, mtDNA content was determined by qPCR. Mitochondrial complex activity and oxidative stress parameters were spectrophotometrically evaluated. Adenine nucleotide levels, adenosine and its metabolites (inosine and hypoxanthine) were determined by reverse-phase HPLC. Pre-diabetic rats showed increased blood glucose levels and impaired glucose tolerance. Both testicular PGC-1α and Sirt3 levels were decreased. NRF-1, NRF-2 and TFAM were not altered. Testicular mtDNA content was decreased. Mitochondrial complex I activity was increased, whereas mitochondrial complex III activity was decreased. Adenylate energy charge was decreased in pre-diabetic rats, as were ATP and ADP levels. Conversely, AMP levels were increased, evidencing a decreased ATP/AMP ratio. Concerning to oxidative stress pre-diabetes decreased testicular antioxidant capacity and increased lipid and protein oxidation. In sum, pre-diabetes compromises testicular mitochondrial function by repressing PGC-1α/Sirt3 axis and mtDNA copy number, declining respiratory capacity and increasing oxidative stress. This study gives new insights into overall testicular bioenergetics at this prodromal stage of disease.
Collapse
Affiliation(s)
- Luís Rato
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal; CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Ana I Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Gonçalo D Tomás
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Maria S Santos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Life Sciences Department, Faculty of Sciences and Technology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Paula I Moreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sílvia Socorro
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - José E Cavaco
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Marco G Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Pedro F Oliveira
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| |
Collapse
|
47
|
Lopes C, Ribeiro M, Duarte AI, Humbert S, Saudou F, Pereira de Almeida L, Hayden M, Rego AC. IGF-1 intranasal administration rescues Huntington's disease phenotypes in YAC128 mice. Mol Neurobiol 2013; 49:1126-42. [PMID: 24347322 DOI: 10.1007/s12035-013-8585-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/04/2013] [Indexed: 02/06/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant disease caused by an expansion of CAG repeats in the gene encoding for huntingtin. Brain metabolic dysfunction and altered Akt signaling pathways have been associated with disease progression. Nevertheless, conflicting results persist regarding the role of insulin-like growth factor-1 (IGF-1)/Akt pathway in HD. While high plasma levels of IGF-1 correlated with cognitive decline in HD patients, other data showed protective effects of IGF-1 in HD striatal neurons and R6/2 mice. Thus, in the present study, we investigated motor phenotype, peripheral and central metabolic profile, and striatal and cortical signaling pathways in YAC128 mice subjected to intranasal administration of recombinant human IGF-1 (rhIGF-1) for 2 weeks, in order to promote IGF-1 delivery to the brain. We show that IGF-1 supplementation enhances IGF-1 cortical levels and improves motor activity and both peripheral and central metabolic abnormalities in YAC128 mice. Moreover, decreased Akt activation in HD mice brain was ameliorated following IGF-1 administration. Upregulation of Akt following rhIGF-1 treatment occurred concomitantly with increased phosphorylation of mutant huntingtin on Ser421. These data suggest that intranasal administration of rhIGF-1 ameliorates HD-associated glucose metabolic brain abnormalities and mice phenotype.
Collapse
Affiliation(s)
- Carla Lopes
- CNC-Center for Neuroscience and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Silva FSG, Ribeiro MPC, Santos MS, Rocha-Pereira P, Santos-Silva A, Custódio JBA. The antiestrogen 4-hydroxytamoxifen protects against isotretinoin-induced permeability transition and bioenergetic dysfunction of liver mitochondria: comparison with tamoxifen. J Bioenerg Biomembr 2013; 45:383-96. [DOI: 10.1007/s10863-013-9517-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 05/21/2013] [Indexed: 10/26/2022]
|
49
|
González-Burgos E, Duarte AI, Carretero ME, Moreira PI, Gómez-Serranillos MP. Mitochondrial-targeted protective properties of isolated diterpenoids from sideritis spp. in response to the deleterious changes induced by H2O2. JOURNAL OF NATURAL PRODUCTS 2013; 76:933-938. [PMID: 23675610 DOI: 10.1021/np400118d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mitochondrial impairment and oxidative stress are considered widely to be central events in many forms of neurodegenerative disease. The current study has evaluated for the first time the potential protective role of three diterpenoids [andalusol (1), conchitriol (2), and lagascatriol (3)] in response to the deleterious H2O2-induced changes on mitochondrial function. U373-MG human astrocytoma cells and PC12 rat adrenal pheochromocytoma cells were used as models for evaluating the cytoprotective potential of these compounds. In the absence of diterpenoids 1-3, H2O2 compromised mitochondrial function, decreasing mitochondrial membrane potential and ATP levels, increasing caspase-3 activity, and disrupting cytosolic and mitochondrial calcium homeostasis. However, treatment with the diterpenoids, prior to H2O2, prevented these mitochondrial perturbations. In particular, 1 and 3 were the most effective compounds in protecting mitochondrial function against H2O2-induced oxidative stress in U373-MG, whereas all three diterpenoids studied were significantly active against PC12 cells. Since consistent evidence has demonstrated the contribution of H2O2 on both progression and pathological development of several human diseases associated with mitochondrial function and oxidative stress responses, compounds 1-3 are worthy of further investigation.
Collapse
Affiliation(s)
- Elena González-Burgos
- Department of Pharmacology, Faculty of Pharmacy, Complutense University, Madrid, Spain
| | | | | | | | | |
Collapse
|
50
|
Silva AC, Almeida S, Laço M, Duarte AI, Domingues J, Oliveira CR, Januário C, Rego AC. Mitochondrial respiratory chain complex activity and bioenergetic alterations in human platelets derived from pre-symptomatic and symptomatic Huntington's disease carriers. Mitochondrion 2013; 13:801-9. [PMID: 23707479 DOI: 10.1016/j.mito.2013.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 04/17/2013] [Accepted: 05/07/2013] [Indexed: 01/14/2023]
Abstract
Mitochondrial dysfunction has been implicated in Huntington's disease (HD) pathogenesis. We analyzed the activity of mitochondrial complexes (Cx) I-IV, protein levels of selected Cx subunits and adenine nucleotides in platelet mitochondria from pre-symptomatic versus symptomatic HD human carriers and age-matched control individuals. Mitochondrial platelets exhibited reduced activity of citrate synthase in pre-symptomatic and Cx-I in pre-symptomatic and symptomatic HD carriers. Positive correlation between Cx activity and protein subunits was observed for Cx-I in symptomatic HD patient's mitochondria. Moreover, AMP increased in mitochondria from pre-symptomatic HD carriers. Results highlight mitochondrial changes occurring before the onset of HD clinical symptoms.
Collapse
Affiliation(s)
- Ana C Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | | | | | | | | | | | | | | |
Collapse
|