1
|
Ntawubizi M, Mukamuhirwa ML. A reflexion on the oxidative stress and animal welfare: a review. Trop Anim Health Prod 2024; 56:396. [PMID: 39601945 DOI: 10.1007/s11250-024-04238-x] [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: 06/04/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
A wide range of studies have documented the role of oxidative stress in the development of chronic pathological disorders and even in the aging itself. However, its significance to modern animal health and welfare remains neglected. Oxidative stress in biological systems refers to a disturbance in the balance between pro-oxidants and antioxidants in favour of the former, leading to potential damage of biomolecules. In farm animals, oxidative stress may be involved in several pathological conditions, including those that are relevant for animal production and the general welfare of the individuals, resulting in some cases in irreversible losses. The oxidative stress concept and how it may result in disease or be prevented are complex questions with no simple answers and therefore, call professionals for deep reflection, to maintain a high standard of animal welfare and production. The aim of this review was to gather relevant information on the characteristics of pro-oxidants and antioxidant as well as their significance in animal production systems.
Collapse
Affiliation(s)
- Martin Ntawubizi
- School of Veterinary Medicine, University of Rwanda-Nyagatare Campus, P.O. Box 57, Nyagatare, Rwanda.
| | - Marie Louise Mukamuhirwa
- School of Veterinary Medicine, University of Rwanda-Nyagatare Campus, P.O. Box 57, Nyagatare, Rwanda
| |
Collapse
|
2
|
Zhan S, Wu W, Hu J, Liu F, Qiao X, Chen L, Zhou Y. The pathogenicity and regulatory function of temperature-sensitive proteins PscTSP in Pseudofabraea citricarpa under high temperature stress. Int J Biol Macromol 2024; 270:132017. [PMID: 38697438 DOI: 10.1016/j.ijbiomac.2024.132017] [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: 03/27/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Citrus fruit rich in beneficial health-promoting nutrients used for functional foods or dietary supplements production. However, its quality and yield were damaged by citrus target spot. Citrus target spot is a low-temperature fungal disease caused by Pseudofabraea citricarpa, resulting in citrus production reductions and economic losses. In this study, transcriptome and gene knockout mutant analyses were performed on the growth and pathogenicity of P. citricarpa under different temperature conditions to quantify the functions of temperature-sensitive proteins (PscTSP). The optimum growth temperature for P. citricarpa strain WZ1 was 20 °C, while it inhibited or stopped growth above 30 °C and stopped growth below 4 °C or above 30 °C. Certain PscTSP-key genes of P. citricarpa were identified under high temperature stress. qRT-PCR analysis confirmed the expression levels of PscTSPs under high temperature stress. PscTSPs were limited by temperature and deletion of the PscTSP-X gene leads to changes in the integrity of citrus cell walls, osmotic regulation, oxidative stress response, calcium regulation, chitin synthesis, and the pathogenicity of P. citricarpa. These results provide insight into the underlying mechanisms of temperature sensitivity and pathogenicity in P. citricarpa, providing a foundation for developing resistance strategies against citrus target spot disease.
Collapse
Affiliation(s)
- Shuang Zhan
- Citrus Research Institute, Southwest University/Chinese Academy of Agricultural Sciences, Chongqing 400712, China
| | - Wang Wu
- Citrus Research Institute, Southwest University/Chinese Academy of Agricultural Sciences, Chongqing 400712, China
| | - Junhua Hu
- Citrus Research Institute, Southwest University/Chinese Academy of Agricultural Sciences, Chongqing 400712, China; Scientific Observing and Experimental Station of Fruit Tree Science (Southwest Region), Ministry of Agriculture, Chongqing 400712, China.
| | - Fengjiao Liu
- Citrus Research Institute, Southwest University/Chinese Academy of Agricultural Sciences, Chongqing 400712, China
| | - Xinghua Qiao
- Wanzhou District of Chongqing Plant Protection and Fruit Tree Technology Promotion Station, Wanzhou, 404000, China
| | - Li Chen
- Wanzhou District of Chongqing Plant Protection and Fruit Tree Technology Promotion Station, Wanzhou, 404000, China
| | - Yan Zhou
- Citrus Research Institute, Southwest University/Chinese Academy of Agricultural Sciences, Chongqing 400712, China; Scientific Observing and Experimental Station of Fruit Tree Science (Southwest Region), Ministry of Agriculture, Chongqing 400712, China
| |
Collapse
|
3
|
Chen X, Gao M, An S, Zhao L, Han W, Wan W, Chen J, Ma S, Cai W, Cao Y, Ding D, Yang YY, Cheng L, Zheng Y. Enhancing adoptive T cell therapy for solid tumor with cell-surface anchored immune checkpoint inhibitor nanogels. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 45:102591. [PMID: 35907618 DOI: 10.1016/j.nano.2022.102591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 07/10/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
The efficacy of Adoptive Cell Therapy (ACT) for solid tumor is still mediocre. This is mainly because tumor cells can hijack ACT T cells' immune checkpoint pathways to exert immunosuppression in the tumor microenvironment. Immune Checkpoint Inhibitors such as anti-PD-1 (aPD1) can counter the immunosuppression, but the synergizing effects of aPD1 to ACT was still not satisfactory. Here we demonstrate an approach to safely anchor aPD1-formed nanogels onto T cell surface via bio-orthogonal click chemistry before adoptive transfer. The spatial-temporal co-existence of aPD1 with ACT T cells and the responsive drug release significantly improved the treatment outcome of ACT in murine solid tumor model. The average tumor weight of the group treated by cell-surface anchored aPD1 was only 18 % of the group treated by equivalent dose of free aPD1 and T cells. The technology can be broadly applicable in ACTs employing natural or Chimeric Antigen Receptor (CAR) T cells.
Collapse
Affiliation(s)
- Xingye Chen
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Mengqian Gao
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Shan An
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Lei Zhao
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Wenqing Han
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Wenjun Wan
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Jin Chen
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Siqi Ma
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Wenhua Cai
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Yanni Cao
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Dawei Ding
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Yi Yan Yang
- Agency for Science Technology and Research, Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Lifang Cheng
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China.
| | - Yiran Zheng
- College of Pharmaceutical Sciences, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou 215123, PR China.
| |
Collapse
|
4
|
Tang L, Zheng Y, Melo MB, Mabardi L, Castaño AP, Xie YQ, Li N, Kudchodkar SB, Wong HC, Jeng EK, Maus MV, Irvine DJ. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat Biotechnol 2018; 36:707-716. [PMID: 29985479 PMCID: PMC6078803 DOI: 10.1038/nbt.4181] [Citation(s) in RCA: 434] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 06/07/2018] [Indexed: 12/24/2022]
Abstract
Adoptive cell therapy (ACT) with antigen-specific T cells has shown remarkable clinical success, but approaches to safely and effectively augment T cell function, especially in solid tumors, remain of great interest. Here we describe a strategy to “backpack” large quantities of supporting protein drugs on T cells using protein nanogels (NGs) that selectively release these cargos in response to T cell receptor (TCR) activation. We design cell surface-conjugated NGs that respond to an increase in T cell surface reduction potential upon antigen recognition, limiting drug release to sites of antigen encounter such as the tumor microenvironment. Using NGs carrying an IL-15 superagonist complex, we demonstrate that relative to systemic administration of free cytokines, NG delivery selectively expands T cells 16-fold in tumors, and allows at least 8-fold higher doses of cytokine to be administered without toxicity. The improved therapeutic window enables substantially increased tumor clearance by murine T cell and human CAR-T cell therapy in vivo.
Collapse
Affiliation(s)
- Li Tang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yiran Zheng
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mariane Bandeira Melo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Llian Mabardi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
| | - Ana P Castaño
- Cellular Immunotherapy Program, Massachusetts General Hospital (MGH) Cancer Center, Charlestown, Massachusetts, USA
| | - Yu-Qing Xie
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Na Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Hing C Wong
- Altor BioScience Corporation, Miramar, Florida, USA
| | - Emily K Jeng
- Altor BioScience Corporation, Miramar, Florida, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital (MGH) Cancer Center, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Darrell J Irvine
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
5
|
Bharath LP, Cho JM, Park SK, Ruan T, Li Y, Mueller R, Bean T, Reese V, Richardson RS, Cai J, Sargsyan A, Pires K, Anandh Babu PV, Boudina S, Graham TE, Symons JD. Endothelial Cell Autophagy Maintains Shear Stress-Induced Nitric Oxide Generation via Glycolysis-Dependent Purinergic Signaling to Endothelial Nitric Oxide Synthase. Arterioscler Thromb Vasc Biol 2017; 37:1646-1656. [PMID: 28684613 DOI: 10.1161/atvbaha.117.309510] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/19/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Impaired endothelial cell (EC) autophagy compromises shear stress-induced nitric oxide (NO) generation. We determined the responsible mechanism. APPROACH AND RESULTS On autophagy compromise in bovine aortic ECs exposed to shear stress, a decrease in glucose uptake and EC glycolysis attenuated ATP production. We hypothesized that decreased glycolysis-dependent purinergic signaling via P2Y1 (P2Y purinoceptor 1) receptors, secondary to impaired autophagy in ECs, prevents shear-induced phosphorylation of eNOS (endothelial nitric oxide synthase) at its positive regulatory site S1117 (p-eNOSS1177) and NO generation. Maneuvers that restore glucose transport and glycolysis (eg, overexpression of GLUT1 [glucose transporter 1]) or purinergic signaling (eg, addition of exogenous ADP) rescue shear-induced p-eNOSS1177 and NO production in ECs with impaired autophagy. Conversely, inhibiting glucose transport via GLUT1 small interfering RNA, blocking purinergic signaling via ectonucleotidase-mediated ATP/ADP degradation (eg, apyrase), or inhibiting P2Y1 receptors using pharmacological (eg, MRS2179 [2'-deoxy-N6-methyladenosine 3',5'-bisphosphate tetrasodium salt]) or genetic (eg, P2Y1-receptor small interfering RNA) procedures inhibit shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Supporting a central role for PKCδT505 (protein kinase C delta T505) in relaying the autophagy-dependent purinergic-mediated signal to eNOS, we find that (1) shear stress-induced activating phosphorylation of PKCδT505 is negated by inhibiting autophagy, (2) shear-induced p-eNOSS1177 and NO generation are restored in autophagy-impaired ECs via pharmacological (eg, bryostatin) or genetic (eg, constitutively active PKCδ) activation of PKCδT505, and (3) pharmacological (eg, rottlerin) and genetic (eg, PKCδ small interfering RNA) PKCδ inhibition prevents shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Key nodes of dysregulation in this pathway on autophagy compromise were revealed in human arterial ECs. CONCLUSIONS Targeted reactivation of purinergic signaling and PKCδ has strategic potential to restore compromised NO generation in pathologies associated with suppressed EC autophagy.
Collapse
Affiliation(s)
- Leena P Bharath
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jae Min Cho
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Seul-Ki Park
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ting Ruan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Youyou Li
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Robert Mueller
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Tyler Bean
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Van Reese
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Russel S Richardson
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jinjin Cai
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ashot Sargsyan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Karla Pires
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Pon Velayutham Anandh Babu
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Sihem Boudina
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Timothy E Graham
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - J David Symons
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.).
| |
Collapse
|
6
|
Otto AM, Hintermair J, Janzon C. NADH-linked metabolic plasticity of MCF-7 breast cancer cells surviving in a nutrient-deprived microenvironment. J Cell Biochem 2015; 116:822-35. [PMID: 25530451 DOI: 10.1002/jcb.25038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/12/2014] [Indexed: 12/21/2022]
Abstract
Characteristic of the tumor microenvironment are fluctuating gradients of reduced nutrient levels and released lactate. A fundamental issue is how tumor cells modulate their metabolic activity when both glucose and glutamine levels become limiting in the presence of high exogenous lactate. For functional analyses, the activities of pyruvate kinase, lactate dehydrogenase (LDH) and plasma membrane NADH oxidase (NOX) as well as cell growth were measured in breast cancer MCF-7 cells cultured in medium containing various concentrations of these metabolites. After 3 days at glucose concentrations below 2.5 mM, cell number was higher with 0.1 mM than with 1.0 mM glutamine, indicating that the glucose/glutamine balance is important for growth. On the other hand, NOX activity increased with increasing glucose >2.5 mM, but only with low glutamine (0.1 mM). Pyruvate kinase activity also increased, with LDH activity remaining 2-3-fold lower. Here NOX could have a complementary role in reoxidizing NADH for glycolysis. Exogenous lactate supported cell survival at limiting concentrations of glucose and glutamine while increasing NOX and pyruvate kinase activities as well as NADH levels. It is proposed that lactate supports cell survival by fuelling gluconeogenesis and/or the TCA cycle in mitochondria, from where NADH could be shuttled to the cytosol and reoxidized by NOX. Cell survival and the metabolic phenotype are thus interrelated to the dynamics of NADH and plasma membrane NOX activity, which are regulated by the balance of glucose/glutamine levels, in conjunction with lactate in a precarious tumor microenvironment.
Collapse
Affiliation(s)
- Angela M Otto
- Institute of Medical Engineering, Technische Universitaet Muenchen, Munich, Germany; Heinz-Nixdorf-Lehrstuhl für Medizinische Elektronik, Technische Universitaet Muenchen, Munich, Germany
| | | | | |
Collapse
|
7
|
Jo HY, Kim Y, Park HW, Moon HE, Bae S, Kim J, Kim DG, Paek SH. The Unreliability of MTT Assay in the Cytotoxic Test of Primary Cultured Glioblastoma Cells. Exp Neurobiol 2015; 24:235-45. [PMID: 26412973 PMCID: PMC4580751 DOI: 10.5607/en.2015.24.3.235] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 08/30/2015] [Accepted: 08/31/2015] [Indexed: 11/19/2022] Open
Abstract
MTT assay is commonly used to assess the cellular cytotoxicity caused by anticancer drugs in glioblastomas. However, there have been some reports insisting that MTT assay exhibited non-specific intracellular reduction of tetrazolium which led to underestimated results of cytotoxicity. Here, we examine whether or not MTT assay can lead to incorrect information regarding alcohol-induced cytotoxicity on immortalized and primary glioblastoma cells. MTT assay was applied to assess the ethanol-induced cytotoxicity at various ethanol concentrations. The cellular cytotoxicity induced by different doses of ethanol was analyzed and compared through several cytotoxic assays. Ethanol-induced cytotoxicity observed through MTT assay on both cell types was shown to be ethanol dose-dependent below a 3% concentration. However, the cytotoxicity was shown to be markedly underestimated only in primary cells at a 5% concentration. RT-PCR and Western Blot showed increased expressions of pro-apoptotic proteins and decreased expressions of anti-apoptotic proteins in an ethanol dose-dependent manner in both cell types. Furthermore, we present a possible mechanism for the unreliable result of MTT assay. A high concentration of ethanol induces more severe membrane damage and increased intracellular concentration of NADH in primary cells which enhances the nonspecific reduction of tetrazolium salt. Together, our findings demonstrate that the cytotoxicity on primary cells could inaccurately be assessed when detected through MTT assay. Therefore, a careful interpretation is needed when one would analyze the cytotoxic results of MTT assay, and it is suggested that other assays must be accompanied to produce more reliable and accurate cytotoxic results on primary glioblastoma cells.
Collapse
Affiliation(s)
- Hwa Yeon Jo
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Yona Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyo Eun Moon
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Seongtae Bae
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - JinWook Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea. ; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03082, Korea
| |
Collapse
|
8
|
Gray JP, Eisen T, Cline GW, Smith PJS, Heart E. Plasma membrane electron transport in pancreatic β-cells is mediated in part by NQO1. Am J Physiol Endocrinol Metab 2011; 301:E113-21. [PMID: 21505151 PMCID: PMC3129843 DOI: 10.1152/ajpendo.00673.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Plasma membrane electron transport (PMET), a cytosolic/plasma membrane analog of mitochondrial electron transport, is a ubiquitous system of cytosolic and plasma membrane oxidoreductases that oxidizes cytosolic NADH and NADPH and passes electrons to extracellular targets. While PMET has been shown to play an important role in a variety of cell types, no studies exist to evaluate its function in insulin-secreting cells. Here we demonstrate the presence of robust PMET activity in primary islets and clonal β-cells, as assessed by the reduction of the plasma membrane-impermeable dyes WST-1 and ferricyanide. Because the degree of metabolic function of β-cells (reflected by the level of insulin output) increases in a glucose-dependent manner between 4 and 10 mM glucose, PMET was evaluated under these conditions. PMET activity was present at 4 mM glucose and was further stimulated at 10 mM glucose. PMET activity at 10 mM glucose was inhibited by the application of the flavoprotein inhibitor diphenylene iodonium and various antioxidants. Overexpression of cytosolic NAD(P)H-quinone oxidoreductase (NQO1) increased PMET activity in the presence of 10 mM glucose while inhibition of NQO1 by its inhibitor dicoumarol abolished this activity. Mitochondrial inhibitors rotenone, antimycin A, and potassium cyanide elevated PMET activity. Regardless of glucose levels, PMET activity was greatly enhanced by the application of aminooxyacetate, an inhibitor of the malate-aspartate shuttle. We propose a model for the role of PMET as a regulator of glycolytic flux and an important component of the metabolic machinery in β-cells.
Collapse
Affiliation(s)
- Joshua P Gray
- United States Coast Guard Academy, New London, Connecticut, USA
| | | | | | | | | |
Collapse
|
9
|
Del Principe D, Avigliano L, Savini I, Catani MV. Trans-plasma membrane electron transport in mammals: functional significance in health and disease. Antioxid Redox Signal 2011; 14:2289-318. [PMID: 20812784 DOI: 10.1089/ars.2010.3247] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Trans-plasma membrane electron transport (t-PMET) has been established since the 1960s, but it has only been subject to more intensive research in the last decade. The discovery and characterization at the molecular level of its novel components has increased our understanding of how t-PMET regulates distinct cellular functions. This review will give an update on t-PMET, with particular emphasis on how its malfunction relates to some diseases, such as cancer, abnormal cell death, cardiovascular diseases, aging, obesity, neurodegenerative diseases, pulmonary fibrosis, asthma, and genetically linked pathologies. Understanding these relationships may provide novel therapeutic approaches for pathologies associated with unbalanced redox state.
Collapse
Affiliation(s)
- Domenico Del Principe
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy.
| | | | | | | |
Collapse
|
10
|
Agarwal A, Desai NR, Makker K, Varghese A, Mouradi R, Sabanegh E, Sharma R. Effects of radiofrequency electromagnetic waves (RF-EMW) from cellular phones on human ejaculated semen: an in vitro pilot study. Fertil Steril 2009; 92:1318-1325. [DOI: 10.1016/j.fertnstert.2008.08.022] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Revised: 07/31/2008] [Accepted: 08/07/2008] [Indexed: 01/10/2023]
|
11
|
Susceptibility of mantle cell lymphomas to reovirus oncolysis. Leuk Res 2009; 34:100-8. [PMID: 19651440 DOI: 10.1016/j.leukres.2009.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 05/22/2009] [Accepted: 05/25/2009] [Indexed: 12/14/2022]
Abstract
Mantle cell lymphoma (MCL) an incurable B-cell, non-Hodgkin lymphoma (NHL) urgently requires new treatments. We assessed reovirus mediated oncolysis in a panel of human MCL cell lines. In vitro, we found the cytopathic effect of reovirus infection ranged from high to very limited and correlated with levels of Ras activation. In vivo, a single reovirus injection intra-tumorally resulted in complete regression of both the injected and the contra-lateral tumor in a subcutaneous bi-tumor model, in one out of three cell lines tested. Reovirus treatment of MCL seems feasible but will need to be guided by the presence of molecular determinants of reovirus susceptibility.
Collapse
|
12
|
Matkar SS, Wrischnik LA, Hellmann-Blumberg U. Production of hydrogen peroxide and redox cycling can explain how sanguinarine and chelerythrine induce rapid apoptosis. Arch Biochem Biophys 2008; 477:43-52. [PMID: 18555791 DOI: 10.1016/j.abb.2008.05.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2008] [Revised: 04/28/2008] [Accepted: 05/20/2008] [Indexed: 11/18/2022]
Abstract
Sanguinarine and chelerythrine are naturally occurring benzophenanthridines with multiple biological activities. Sanguinarine is believed to be a potential anticancer agent but its mechanism of action has not been fully elucidated. We previously found that it causes oxidative DNA damage and very rapid apoptosis that is not mediated by p53-dependent DNA damage signaling. Here we show that sanguinarine and chelerythrine cause the production of large amounts of reactive oxygen species (ROS), in particular hydrogen peroxide, which may deplete cellular antioxidants and provide a signal for rapid execution of apoptosis. Several oxidoreductases contribute to cell death induced by sanguinarine and chelerythrine which appear to be reduced upon entering the cell. We propose a model in which the generation of lethal amounts of hydrogen peroxide is explained by enzyme-catalyzed redox cycling between the reduced and oxidized forms of the phenanthridines and discuss the implications of such a mechanism for potential pharmaceutical applications.
Collapse
Affiliation(s)
- Smita S Matkar
- Department of Chemistry, University of the Pacific, 3601 Pacific Avenue, Stockton, CA 95211, USA
| | | | | |
Collapse
|
13
|
Park BC, Park SH, Paek SH, Park SY, Kwak MK, Choi HG, Yong CS, Yoo BK, Kim JA. Chloroquine-induced nitric oxide increase and cell death is dependent on cellular GSH depletion in A172 human glioblastoma cells. Toxicol Lett 2008; 178:52-60. [DOI: 10.1016/j.toxlet.2008.02.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Revised: 01/31/2008] [Accepted: 02/06/2008] [Indexed: 01/31/2023]
|
14
|
Flaherty MM, Rish KR, Smith A, Crumbliss AL. An investigation of hemopexin redox properties by spectroelectrochemistry: biological relevance for heme uptake. Biometals 2007; 21:239-48. [PMID: 17712531 DOI: 10.1007/s10534-007-9112-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 07/26/2007] [Indexed: 10/22/2022]
Abstract
Hemopexin (HPX) has two principal roles: it sequesters free heme in vivo for the purpose of preventing the toxic effects of this moiety, which is largely due to heme's ability to catalyze free radical formation, and it transports heme intracellularly thus limiting its availability as an iron source for pathogens. Spectroelectrochemistry was used to determine the redox potential for heme and meso-heme (mH) when bound by HPX. At pH 7.2, the heme-HPX assembly exhibits E (1/2) values in the range 45-90 mV and the mH-HPX assembly in the range 5-55 mV, depending on environmental electrolyte identity. The E (1/2) value exhibits a 100 mV positive shift with a change in pH from 7.2 to 5.5 for mH-HPX, suggesting a single proton dependent equilibrium. The E (1/2) values for heme-HPX are more positive in the presence of NaCl than KCl indicating that Na(+), as well as low pH (5.5) stabilizes ferro-heme-HPX. Furthermore, comparing KCl with K(2)HPO(4), the chloride salt containing system has a lower potential, indicating that heme-HPX is easier to oxidize. These physical properties related to ferri-/ferro-heme reduction are both structurally and biologically relevant for heme release from HPX for transport and regulation of heme oxygenase expression. Consistent with this, when the acidification of endosomes is prevented by bafilomycin then heme oxygenase-1 induction by heme-HPX no longer occurs.
Collapse
Affiliation(s)
- Meghan M Flaherty
- Department of Chemistry, Duke University, Box 90346, Durham, NC 27708-0346, USA
| | | | | | | |
Collapse
|
15
|
Herst PM, Petersen T, Jerram P, Baty J, Berridge MV. The antiproliferative effects of phenoxodiol are associated with inhibition of plasma membrane electron transport in tumour cell lines and primary immune cells. Biochem Pharmacol 2007; 74:1587-95. [PMID: 17904534 DOI: 10.1016/j.bcp.2007.08.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 08/15/2007] [Accepted: 08/15/2007] [Indexed: 12/19/2022]
Abstract
Although the redox-active synthetic isoflavene, phenoxodiol, is in Phase 3 clinical trials for drug-resistant ovarian cancer, and in early stage clinical trials for prostate and cervical cancer, its primary molecular target is unknown. Nevertheless, phenoxodiol inhibits proliferation of many cancer cell lines and induces apoptosis by disrupting FLICE-inhibitory protein, FLIP, expression and by caspase-dependent and -independent degradation of the X-linked inhibitor of apoptosis, XIAP. In addition, phenoxodiol sensitizes drug-resistant tumour cells to anticancer drugs including paclitaxel, carboplatin and gemcitabine. Here, we investigate the effects of phenoxodiol on plasma membrane electron transport (PMET) and cell proliferation in human leukemic HL60 cells and mitochondrial gene knockout HL60rho(o) cells that exhibit elevated PMET. Phenoxodiol inhibited PMET by both HL60 (IC(50) 32 microM) and HL60rho(o) (IC(50) 70 microM) cells, and this was associated with inhibition of cell proliferation (IC(50) of 2.8 and 6.7 microM, respectively), pan-caspase activation and apoptosis. Unexpectedly, phenoxodiol also inhibited PMET by activated murine splenic T cells (IC(50) of 29 microM) as well as T cell proliferation (IC(50) of 2.5 microM). In contrast, proliferation of WI-38 cells and HUVECs was only weakly affected by phenoxodiol. These results indicate that PMET may be a primary target for phenoxodiol in tumour cells and in activated T cells.
Collapse
Affiliation(s)
- P M Herst
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6005, New Zealand
| | | | | | | | | |
Collapse
|
16
|
Rish KR, Swartzlander R, Sadikot TN, Berridge MV, Smith A. Interaction of heme and heme-hemopexin with an extracellular oxidant system used to measure cell growth-associated plasma membrane electron transport. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2007; 1767:1107-17. [PMID: 17643387 DOI: 10.1016/j.bbabio.2007.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Revised: 06/12/2007] [Accepted: 06/13/2007] [Indexed: 11/18/2022]
Abstract
Since redox active metals are often transported across membranes into cells in the reduced state, we have investigated whether exogenous ferri-heme or heme bound to hemopexin (HPX), which delivers heme to cells via receptor-mediated endocytosis, interact with a cell growth-associated plasma membrane electron transport (PMET) pathway. PMET reduces the cell-impermeable tetrazolium salt, WST-1, in the presence of the mandatory low potential intermediate electron acceptor, mPMS. In human promyelocytic (HL60) cells, protoheme (iron protoporphyrin IX; 2,4-vinyl), mesoheme (2,4-ethyl) and deuteroheme (2,4-H) inhibited reduction of WST-1/mPMS in a saturable manner supporting interaction with a finite number of high affinity acceptor sites (Kd 221 nM for naturally occurring protoheme). A requirement for the redox-active iron was shown using gallium-protoporphyrin IX (PPIX) and tin-PPIX. Heme-hemopexin, but not apo-hemopexin, also inhibited WST-1 reduction, and copper was required. Importantly, since neither heme nor heme-hemopexin replace mPMS as an intermediate electron acceptor and since inhibition of WST-1/mPMS reduction requires living cells, the experimental evidence supports the view that heme and heme-hemopexin interact with electrons from PMET. We therefore propose that heme and heme-hemopexin are natural substrates for this growth-associated electron transfer across the plasma membrane.
Collapse
Affiliation(s)
- Kimberly R Rish
- School of Biological Sciences, University of Missouri-Kansas City, 5007 Rockhill Road, Kansas City, MO 64110-2499, USA
| | | | | | | | | |
Collapse
|
17
|
Rajwa B, Bernas T, Acker H, Dobrucki J, Robinson JP. Single- and two-photon spectral imaging of intrinsic fluorescence of transformed human hepatocytes. Microsc Res Tech 2007; 70:869-79. [PMID: 17661363 DOI: 10.1002/jemt.20497] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Autofluorescence (AF) originating from the cytoplasmic region of mammalian cells has been thoroughly investigated; however, AF from plasma membranes of viable intact cells is less well known, and has been mentioned only in a few older publications. Herein, we report results describing single- and two-photon spectral properties of a strong yellowish-green AF confined to the plasma-membrane region of transformed human hepatocytes (HepG2) grown in vitro as small three-dimensional aggregates or as monolayers. The excitation-emission characteristics of the membrane AF indicate that it may originate from a flavin derivative. Furthermore, the AF was closely associated with the plasma membranes of HepG2 cells, and its presence and intensity were dependent on cell metabolic state, membrane integrity and presence of reducing agents. This AF could be detected both in live intact cells and in formaldehyde-fixed cells.
Collapse
Affiliation(s)
- Bartek Rajwa
- Purdue University Cytometry Laboratories, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana 47907-2057, USA.
| | | | | | | | | |
Collapse
|
18
|
Herst PM, Berridge MV. Cell surface oxygen consumption: a major contributor to cellular oxygen consumption in glycolytic cancer cell lines. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2006; 1767:170-7. [PMID: 17266920 DOI: 10.1016/j.bbabio.2006.11.018] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2006] [Revised: 11/23/2006] [Accepted: 11/29/2006] [Indexed: 01/09/2023]
Abstract
Oxygen consumption for bioenergetic purposes has long been thought to be the prerogative of mitochondria. Nevertheless, mitochondrial gene knockout (rho(0)) cells that are defective in mitochondrial respiration require oxygen for growth and consume oxygen at the cell surface via trans-plasma membrane electron transport (tPMET). This raises the possibility that cell surface oxygen consumption may support glycolytic energy metabolism by reoxidising cytosolic NADH to facilitate continued glycolysis. In this paper we determined the extent of cell surface oxygen consumption in a panel of 19 cancer cell lines. Non-mitochondrial (myxothiazol-resistant) oxygen consumption was demonstrated to consist of at least two components, cell surface oxygen consumption (inhibited by extracellular NADH) and basal oxygen consumption (insensitive to both myxothiazol and NADH). The extent of cell surface oxygen consumption varied considerably between parental cell lines from 1% to 80% of total oxygen consumption rates. In addition, cell surface oxygen consumption was found to be associated with low levels of superoxide production and to contribute significantly (up to 25%) to extracellular acidification in HL60rho(0) cells. In summary, cell surface oxygen consumption contributes significantly to total cellular oxygen consumption, not only in rho(0) cells but also in mitochondrially competent tumour cell lines with glycolytic metabolism.
Collapse
Affiliation(s)
- Patries M Herst
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington, New Zealand.
| | | |
Collapse
|
19
|
Knight SAB, Dancis A. Reduction of 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide inner salt (XTT) is dependent on CaFRE10 ferric reductase for Candida albicans grown in unbuffered media. MICROBIOLOGY-SGM 2006; 152:2301-2308. [PMID: 16849796 DOI: 10.1099/mic.0.28843-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The reduction of 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide inner salt (XTT) and other tetrazolium salts is widely used as an assay for bacterial, fungal and mammalian cell viability, but the genes encoding the reductase activities have not been defined. Here, it was shown that XTT and plasma membrane ferric reductase activities were 10-40-fold greater in Candida albicans than in Saccharomyces cerevisiae. XTT reductase activity was induced fivefold in C. albicans grown in low-iron conditions compared with iron-replete conditions, and for cells grown in unbuffered (pH 4.0-4.4) medium, XTT reductase activity was largely dependent on CaFRE10. XTT reductase activity of C. albicans grown in medium buffered to pH 6.8 was independent of CaFRE10 but, nonetheless, was upregulated in cells deprived of iron. Reduction of 2-(4,5-dimethyl-2-thiazolyl)-3,5-diphenyl-2H-tetrazolium bromide (MTT), a membrane-permeable tetrazolium salt, occurred at an intracellular location and was independent of CaFRE10. However, MTT activity was induced by iron deprivation in C. albicans but not in S. cerevisiae. C. albicans possessed multiple iron- and pH-regulated reductase activities capable of reducing tetrazolium salts, but, when grown in unbuffered medium, CaFRE10 was required for XTT reductase activity.
Collapse
Affiliation(s)
- Simon A B Knight
- University of Pennsylvania School of Medicine, Department of Medicine, Division of Hematology/Oncology, 730 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6160, USA
| | - Andrew Dancis
- University of Pennsylvania School of Medicine, Department of Medicine, Division of Hematology/Oncology, 730 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6160, USA
| |
Collapse
|
20
|
Kennett EC, Kuchel PW. Plasma membrane oxidoreductases: effects on erythrocyte metabolism and redox homeostasis. Antioxid Redox Signal 2006; 8:1241-7. [PMID: 16910771 DOI: 10.1089/ars.2006.8.1241] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Plasma membrane oxidoreductases (PMORs) have been found in the membranes of all cells. These systems have been studied extensively in the human erythrocyte, so much is known about their activity and effect on erythrocyte cellular functioning. PMORs have been shown to be involved in a number of events associated with cell growth and function in other cell lines, but perhaps their most important role, especially in the nucleus- free mature erythrocyte, is as a redox sensor. The PMOR reduces extracellular oxidants by using the reducing power of intracellular antioxidants, making the cell metabolism respond to changes in the local redox environment. Thus, the activity of the PMOR is closely linked to the metabolic status of the erythrocyte. The main intracellular reductant for this system is ascorbic acid; however, the cell must also have the ability to supply NADH for full activity. Nuclear magnetic resonance studies on the effects of extracellular oxidants on intracellular metabolism have increased our knowledge of the intimate link between PMOR activity and metabolism, and these studies are reviewed here in detail.
Collapse
Affiliation(s)
- Eleanor C Kennett
- School of Molecular and Microbial Biosciences, University of Sydney, NSW, Australia
| | | |
Collapse
|
21
|
Martirosyan A, Leonard S, Shi X, Griffith B, Gannett P, Strobl J. Actions of a histone deacetylase inhibitor NSC3852 (5-nitroso-8-quinolinol) link reactive oxygen species to cell differentiation and apoptosis in MCF-7 human mammary tumor cells. J Pharmacol Exp Ther 2006; 317:546-52. [PMID: 16497787 DOI: 10.1124/jpet.105.096891] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
NSC3852 (5-nitroso-8-quinolinol) has cell differentiation and antiproliferative activity in human breast cancer cells in tissue culture and antitumor activity in mice bearing P388 and L1210 leukemic cells. We investigated the mechanism of NSC3852 action in MCF-7 human breast cancer cells using electron spin resonance (ESR). Reactive oxygen species (ROS) were detected in MCF-7 cell suspensions incubated with NSC3852 using the spin trap 5,5-dimethyl-1-pyrroline-N-oxide (DMPO). Formation of the DMPO-OH adduct was quenched by the addition of superoxide dismutase but not by catalase, and we concluded that superoxide was generated in the NSC3852-treated cells. The flavoprotein inhibitor diphenylene iodonium suppressed ROS production, providing evidence for the involvement of a flavin-dependent enzyme system in the ROS response to NSC3852. A biologically significant oxidative response to NSC3852 occurred in MCF-7 cells. An early marker of oxidative stress was a decrease in the [glutathione]/[glutathione disulfide] ratio 1 h after NSC3852 addition. Oxidative DNA damage, marked by the presence of 8-oxoguanine, and DNA-strand breakage occurred in cells exposed to NSC3852 for 24 h. Apoptosis peaked 48 h after exposure to NSC3852. Pretreatment with the glutathione precursor N-acetyl-l-cysteine (NAC) prevented DNA-strand breakage and apoptosis. Pretreatment with NAC also reversed NSC3852 decreases in E2F1, Myc, and phosphorylated retinoblastoma protein, indicative of redox-sensitive pathway(s) in MCF-7 cells during G(1) phase of the cell cycle. We conclude that ROS formation is involved in the apoptotic and cell differentiation responses to NSC3852 in MCF-7 cells.
Collapse
Affiliation(s)
- Anna Martirosyan
- Department of Biochemistry and Molecular Pharmacology, West Virginia University, Morgantown, West Virginia, USA
| | | | | | | | | | | |
Collapse
|
22
|
Baker MA, Aitken RJ. Reactive oxygen species in spermatozoa: methods for monitoring and significance for the origins of genetic disease and infertility. Reprod Biol Endocrinol 2005; 3:67. [PMID: 16313680 PMCID: PMC1315356 DOI: 10.1186/1477-7827-3-67] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 11/29/2005] [Indexed: 11/10/2022] Open
Abstract
Human spermatozoa generate low levels of reactive oxygen species in order to stimulate key events, such as tyrosine phosphorylation, associated with sperm capacitation. However, if the generation of these potentially pernicious oxygen metabolites becomes elevated for any reason, spermatozoa possess a limited capacity to protect themselves from oxidative stress. As a consequence, exposure of human spermatozoa to intrinsically- or extrinsically- generated reactive oxygen intermediates can result in a state of oxidative stress characterized by peroxidative damage to the sperm plasma membrane and DNA damage to the mitochondrial and nuclear genomes. Oxidative stress in the male germ line is associated with poor fertilization rates, impaired embryonic development, high levels of abortion and increased morbidity in the offspring, including childhood cancer. In this review, we consider the possible origins of oxidative damage to human spermatozoa and reflect on the important contribution such stress might make to the origins of genetic disease in our species.
Collapse
Affiliation(s)
- Mark A Baker
- The ARC Centre of Excellence in Biotechnology and Development, Reproductive Science Group, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - R John Aitken
- The ARC Centre of Excellence in Biotechnology and Development, Reproductive Science Group, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
23
|
Reddy Pagala V, Park J, Reed DW, Hartzell PL. Cellular localization of D-lactate dehydrogenase and NADH oxidase from Archaeoglobus fulgidus. ARCHAEA-AN INTERNATIONAL MICROBIOLOGICAL JOURNAL 2005; 1:95-104. [PMID: 15803647 PMCID: PMC2685561 DOI: 10.1155/2002/297264] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Members of the genus Archaeoglobus are hyperthermophilic sulfate reducers with an optimal growth temperature of 83 degrees C. Archaeoglobus fulgidus can utilize simple compounds including D-lactate, L-lactate and pyruvate as the sole substrate for carbon and electrons for dissimilatory sulfate reduction. Previously we showed that this organism makes a D-lactate dehydrogenase (Dld) that requires FAD and Zn2+ for activity. To determine the cellular location and topology of Dld and to identify proteins that interact with Dld, an antibody directed against Dld was prepared. Immunocytochemical studies using gold particle-coated secondary antibodies show that more than 85% of Dld is associated with the membrane. A truncated form of Dld was detected in immunoblots of whole cells treated with protease, showing that Dld is an integral membrane protein and that a significant portion of Dld, including part of the FAD-binding pocket, is outside the membrane facing the S-layer. The gene encoding Dld is part of an operon that includes noxA2, which encodes one of several NADH oxidases in A. fulgidus. Previous studies have shown that NoxA2 remains bound to Dld during purification. Thin sections of A. fulgidus probed simultaneously with antibodies against Dld and NoxA2 show that both proteins co-localized to the same sites in the membrane. Although these data show a tight interaction between NoxA2 and Dld, the role of NoxA2 in electron transport reactions is unknown. Rather, NoxA2 may protect proteins involved in electron transfer by reducing O2 to H2O2 or H2O.
Collapse
Affiliation(s)
- Vishwajeeth Reddy Pagala
- Department of Microbiology, Molecular Biology, and Biochemistry, University of Idaho, Moscow, ID 83844-3052, USA
| | - Joohye Park
- Department of Microbiology, Molecular Biology, and Biochemistry, University of Idaho, Moscow, ID 83844-3052, USA
| | - David W. Reed
- Department of Microbiology, Molecular Biology, and Biochemistry, University of Idaho, Moscow, ID 83844-3052, USA
- Idaho National Engineering and Environmental Laboratories, Idaho Falls, ID 83415, USA
| | - Patricia L. Hartzell
- 142 Life Science, University of Idaho, Moscow, ID 83844-3052, USA
- Corresponding author ()
| |
Collapse
|
24
|
Scarlett DJG, Herst PM, Berridge MV. Multiple proteins with single activities or a single protein with multiple activities: the conundrum of cell surface NADH oxidoreductases. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2005; 1708:108-19. [PMID: 15882838 DOI: 10.1016/j.bbabio.2005.03.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2004] [Revised: 03/13/2005] [Accepted: 03/14/2005] [Indexed: 10/25/2022]
Abstract
Reduction of the cell-impermeable tetrazolium salt WST-1 has been used to characterise two plasma membrane NADH oxidoreductase activities in human cells. The trans activity, measured with WST-1 and the intermediate electron acceptor mPMS, utilises reducing equivalents from intracellular sources, while the surface activity, measured with WST-1 and extracellular NADH, is independent of intracellular metabolism. Whether these two activities involve distinct proteins or are inherent to a single protein is unclear. In this work, we have attempted to address this question by examining the relationship between the trans and surface WST-1-reducing activities and a third well-characterised family of cell surface oxidases, the ECTO-NOX proteins. Using blue native-polyacrylamide gel electrophoresis, we have identified a complex in the plasma membranes of human 143B osteosarcoma cells responsible for the NADH-dependent reduction of WST-1. The dye-reducing activity of the 300 kDa complex was attributed to a 70 kDa NADH oxidoreductase activity that cross-reacted with antisera against the ECTO-NOX protein CNOX. Differences in enzyme activities and inhibitor profiles between the WST-1-reducing NADH oxidoreductase enzyme in the presence of NADH or mPMS and the ECTO-NOX family are reconciled in terms of the different purification methods and assay systems used to study these proteins.
Collapse
|
25
|
Berridge MV, Herst PM, Tan AS. Tetrazolium dyes as tools in cell biology: new insights into their cellular reduction. BIOTECHNOLOGY ANNUAL REVIEW 2005; 11:127-52. [PMID: 16216776 DOI: 10.1016/s1387-2656(05)11004-7] [Citation(s) in RCA: 1358] [Impact Index Per Article: 67.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Tetrazolium salts have become some of the most widely used tools in cell biology for measuring the metabolic activity of cells ranging from mammalian to microbial origin. With mammalian cells, fractionation studies indicate that the reduced pyridine nucleotide cofactor, NADH, is responsible for most MTT reduction and this is supported by studies with whole cells. MTT reduction is associated not only with mitochondria, but also with the cytoplasm and with non-mitochondrial membranes including the endosome/lysosome compartment and the plasma membrane. The net positive charge on tetrazolium salts like MTT and NBT appears to be the predominant factor involved in their cellular uptake via the plasma membrane potential. However, second generation tetrazolium dyes that form water-soluble formazans and require an intermediate electron acceptor for reduction (XTT, WST-1 and to some extent, MTS), are characterised by a net negative charge and are therefore largely cell-impermeable. Considerable evidence indicates that their reduction occurs at the cell surface, or at the level of the plasma membrane via trans-plasma membrane electron transport. The implications of these new findings are discussed in terms of the use of tetrazolium dyes as indicators of cell metabolism and their applications in cell biology.
Collapse
Affiliation(s)
- Michael V Berridge
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, New Zealand.
| | | | | |
Collapse
|
26
|
Vergani L, Floreani M, Russell A, Ceccon M, Napoli E, Cabrelle A, Valente L, Bragantini F, Leger B, Dabbeni-Sala F. Antioxidant defences and homeostasis of reactive oxygen species in different human mitochondrial DNA-depleted cell lines. ACTA ACUST UNITED AC 2004; 271:3646-56. [PMID: 15355341 DOI: 10.1111/j.1432-1033.2004.04298.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Three pairs of parental (rho+) and established mitochondrial DNA depleted (rho0) cells, derived from bone, lung and muscle were used to verify the influence of the nuclear background and the lack of efficient mitochondrial respiratory chain on antioxidant defences and homeostasis of intracellular reactive oxygen species (ROS). Mitochondrial DNA depletion significantly lowered glutathione reductase activity, glutathione (GSH) content, and consistently altered the GSH2 : oxidized glutathione ratio in all of the rho0 cell lines, albeit to differing extents, indicating the most oxidized redox state in bone rho0 cells. Activity, as well as gene expression and protein content, of superoxide dismutase showed a decrease in bone and muscle rho0 cell lines but not in lung rho0 cells. GSH peroxidase activity was four times higher in all three rho0 cell lines in comparison to the parental rho+, suggesting that this may be a necessary adaptation for survival without a functional respiratory chain. Taken together, these data suggest that the lack of respiratory chain prompts the cells to reduce their need for antioxidant defences in a tissue-specific manner, exposing them to a major risk of oxidative injury. In fact bone-derived rho0 cells displayed the highest steady-state level of intracellular ROS (measured directly by 2',7'-dichlorofluorescin, or indirectly by aconitase activity) compared to all the other rho+ and rho0 cells, both in the presence or absence of glucose. Analysis of mitochondrial and cytosolic/iron regulatory protein-1 aconitase indicated that most ROS of bone rho0 cells originate from sources other than mitochondria.
Collapse
Affiliation(s)
- Lodovica Vergani
- Dipartimento di Scienze Neurologiche, Universita di Padova, Padova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Moldovan L, Moldovan NI. Oxygen free radicals and redox biology of organelles. Histochem Cell Biol 2004; 122:395-412. [PMID: 15452718 DOI: 10.1007/s00418-004-0676-y] [Citation(s) in RCA: 300] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2004] [Indexed: 10/26/2022]
Abstract
The presence and supposed roles of reactive oxygen species (ROS) were reported in literature in a myriad of instances. However, the breadth and depth of their involvement in cellular physiology and pathology, as well as their relationship to the redox environment can only be guessed from specialized reports. Whatever their circumstances of formation or consequences, ROS seem to be conspicuous components of intracellular milieu. We sought to verify this assertion, by collecting the available evidence derived from the most recent publications in the biomedical field. Unlike other reviews with similar objectives, we centered our analysis on the subcellular compartments, namely on organelles, grouped according to their major functions. Thus, plasma membrane is a major source of ROS through NAD(P)H oxidases located on either side. Enzymes of the same class displaying low activity, as well as their components, are also present free in cytoplasm, regulating the actin cytoskeleton and cell motility. Mitochondria can be a major source of ROS, mainly in processes leading to apoptosis. The protein synthetic pathway (endoplasmic reticulum and Golgi apparatus), including the nucleus, as well as protein turnover, are all exquisitely sensitive to ROS-related redox conditions. The same applies to the degradation pathways represented by lysosomes and peroxisomes. Therefore, ROS cannot be perceived anymore as a mere harmful consequence of external factors, or byproducts of altered cellular metabolism. This may explain why the indiscriminate use of anti-oxidants did not produce the expected "beneficial" results in many medical applications attempted so far, underlying the need for a deeper apprehension of the biological roles of ROS, particularly in the context of the higher cellular order of organelles.
Collapse
Affiliation(s)
- Leni Moldovan
- Davis Heart and Lung Research Institute, Room. 305D, The Ohio State University, 473 W 12th Avenue, Columbus, OH 43210, USA.
| | | |
Collapse
|
28
|
Herst PM, Tan AS, Scarlett DJG, Berridge MV. Cell surface oxygen consumption by mitochondrial gene knockout cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2004; 1656:79-87. [PMID: 15178469 DOI: 10.1016/j.bbabio.2004.01.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2003] [Revised: 01/29/2004] [Accepted: 01/30/2004] [Indexed: 11/18/2022]
Abstract
Mitochondrial gene knockout (rho(0)) cells that depend on glycolysis for their energy requirements show an increased ability to reduce cell-impermeable tetrazolium dyes by electron transport across the plasma membrane. In this report, we show for the first time, that oxygen functions as a terminal electron acceptor for trans-plasma membrane electron transport (tPMET) in HL60rho(0) cells, and that this cell surface oxygen consumption is associated with oxygen-dependent cell growth in the absence of mitochondrial electron transport function. Non-mitochondrial oxygen consumption by HL60rho(0) cells was extensively inhibited by extracellular NADH and NADPH, but not by NAD(+), localizing this process at the cell surface. Mitochondrial electron transport inhibitors and the uncoupler, FCCP, did not affect oxygen consumption by HL60rho(0) cells. Inhibitors of glucose uptake and glycolysis, the ubiquinone redox cycle inhibitors, capsaicin and resiniferatoxin, the flavin centre inhibitor, diphenyleneiodonium, and the NQO1 inhibitor, dicoumarol, all inhibited oxygen consumption by HL60rho(0) cells. Similarities in inhibition profiles between non-mitochondrial oxygen consumption and reduction of the cell-impermeable tetrazolium dye, WST-1, suggest that both systems may share a common tPMET pathway. This is supported by the finding that terminal electron acceptors from both pathways compete for electrons from intracellular NADH.
Collapse
Affiliation(s)
- Patries M Herst
- Department of Radiation Therapy, Wellington School of Medicine and Health Sciences, University of Otago, PO Box 7343, Wellington, New Zealand
| | | | | | | |
Collapse
|
29
|
Aitken RJ, Ryan AL, Baker MA, McLaughlin EA. Redox activity associated with the maturation and capacitation of mammalian spermatozoa. Free Radic Biol Med 2004; 36:994-1010. [PMID: 15059640 DOI: 10.1016/j.freeradbiomed.2004.01.017] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2003] [Revised: 12/10/2003] [Accepted: 01/23/2004] [Indexed: 11/29/2022]
Abstract
As rat spermatozoa undergo epididymal maturation, they acquire the ability to exhibit a spontaneous burst of luminol-peroxidase-dependent chemiluminescence when released into a simple, defined culture medium. This activity was suppressed by inhibitors of plasma membrane redox systems such as diphenylene iodonium, p-chloromercuribenzenesulfonic acid, and capsaicin, but was resistant to inhibition by resiniferatoxin and rotenone. The luminol-peroxidase signal was dependent on the presence of bicarbonate, enhanced by the substitution of fructose for glucose, and severely suppressed by desferoxamine, superoxide dimutase, and catalase. Both L- and D-arginine were stimulatory, suggesting the involvement of *NO in this spontaneous chemiluminescence activity. The L-arginine-dependent, but not the D-arginine-dependent, activity was significantly suppressed by an inhibitor of nitric oxide synthase (N(G)-nitro-L-arginine methyl ester). L- and D-arginine could also stimulate redox activity observed in immature caput epididymal cells, but only after prolonged incubation. The inhibitory effects of uric acid and ascorbate suggested the chemiluminescence signal might be induced by peroxynitrite. This conclusion was supported by confocal imaging of the cells following treatment with 4-amino-5-methylamino-2',7'-difluorofluorescein. Stimulation or suppression of the redox activity detected by luminol-peroxidase led to corresponding changes in the ability of the spermatozoa to exhibit acrosomal exocytosis, indicating that this pathway is of fundamental biological significance.
Collapse
Affiliation(s)
- R John Aitken
- ARC Centre of Excellence in Biotechnology and Development, Discipline of Biological Sciences, University of Newcastle, New South Wales, Australia.
| | | | | | | |
Collapse
|
30
|
Scarlett DJ, Herst P, Tan A, Prata C, Berridge M. Mitochondrial gene-knockout (rho0) cells: a versatile model for exploring the secrets of trans-plasma membrane electron transport. Biofactors 2004; 20:199-206. [PMID: 15706057 DOI: 10.1002/biof.5520200404] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Plasma membrane electron transport (tPMET) pathways have been identified in all living cells, and a wide variety of tools have been used to study these processes. In our laboratory we have used the cell-impermeable tetrazolium dye WST-1, together with the mitochondrial gene knockout (rho0) cell model, to investigate one of these pathways. We have shown that growth of HL60rho0 cells is dependent on oxygen, and that these cells consume oxygen at the cell surface. Similarities in inhibition profiles between non-mitochondrial oxygen consumption and WST-1 reduction suggest that both systems share a common tPMET pathway. In support of this, oxygen was shown to compete with the intermediate electron acceptor that mediates WST-1 reduction, for reducing electrons. The observation that tPMET activity is higher in rho0 cells compared to their mitochondrially-competent counterparts was shown to be the result of competition between the mitochondrial and plasma membrane electron transport systems for intracellular reducing equivalents. Elevated rates of dye reduction appear to be mediated through increased expression of the key components of tPMET, which include the cell surface NADH oxidase, CNOX. These findings have played a critical role in shaping our current understanding of the mechanisms of this particular pathway of tPMET.
Collapse
|
31
|
Zhang DX, Zou AP, Li PL. Ceramide-induced activation of NADPH oxidase and endothelial dysfunction in small coronary arteries. Am J Physiol Heart Circ Physiol 2003; 284:H605-12. [PMID: 12424096 DOI: 10.1152/ajpheart.00697.2002] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We tested the hypothesis that ceramide induces endothelial dysfunction in small coronary arteries via NADPH oxidase-mediated superoxide and resulting peroxynitrite formation. With the use of dihydroethidium as a superoxide indicator, C(2)-ceramide was found to increase superoxide production in the endothelial cells of small coronary arteries, which was inhibited by the NADPH oxidase inhibitors N-vanillylnonanamide, apocynin, and diphenylene iodonium. NADPH oxidase expression was confirmed in endothelial cells, as indicated by the immunoblotting of its subunits gp91(phox) and p47(phox). C(2)-ceramide increased NADPH oxidase activity by 52%, which was blocked by NADPH oxidase inhibitors but not by inhibitors of NO synthase, xanthine oxidase, and mitochondrial electron transport chain enzymes. By Western blot analysis, ceramide-induced NADPH oxidase activation was found to be associated with the translocation of p47(phox) to the membrane. In isolated and pressurized small coronary arteries, N-vanillylnonanamide, apocynin, or uric acid, a peroxynitrite scavenger, largely restored the inhibitory effects of ceramide on bradykinin- and A-23187-induced vasorelaxation. With the use of nitrotyrosine as a marker, C(2)-ceramide was found to increase peroxynitrite in small coronary arteries, which could be blocked by uric acid. We conclude that NADPH oxidase-mediated superoxide production and subsequent peroxynitrite formation mediate ceramide-induced endothelial dysfunction in small coronary arteries.
Collapse
Affiliation(s)
- David X Zhang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee 53226, USA
| | | | | |
Collapse
|
32
|
Abstract
Radical attack on proteins in the presence of O(2) gives protein hydroperoxides in high yields. These peroxides are decomposed by transition metal ions, reducing agents, UV light and heat, with the formation of a range of reactive radicals that are capable of initiating further damage. Evidence has been presented for the formation of alcohols as stable products of peroxide decomposition, and these have been employed as markers of oxidative damage in vivo. The mechanism of formation of these alcohols is unclear, with both radical and nonradical pathways capable of generating these products. In this study we have investigated the reduction of peptide and protein hydroperoxides by THP-1 (human monocyte-like) cells and it is shown that this process is accompanied by radical formation as detected by EPR spin trapping. The radicals detected, which are similar to those detected from metal-ion catalyzed reduction, are generated externally to the cell. In the absence of cells, or with cell-conditioned media or cell lysates, lower concentrations of radicals were detected, indicating that intact cells are required for rapid hydroperoxide decomposition. The rate of radical generation was enhanced by preloading the cells with ascorbate, and this was accompanied by intracellular formation of the ascorbate radical. It is proposed that decomposition of some amino acid and peptide hydroperoxides occurs extracellularly via the involvement of a cell-surface reducing system, such as a trans-plasma membrane electron transport system (TPMET) either directly, or indirectly via redox cycling of trace transition metal ions.
Collapse
|
33
|
Wright MV, Kuhn TB. CNS neurons express two distinct plasma membrane electron transport systems implicated in neuronal viability. J Neurochem 2002; 83:655-64. [PMID: 12390527 DOI: 10.1046/j.1471-4159.2002.01176.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Trans-plasma membrane electron transport is critical for maintaining cellular redox balance and viability, yet few, if any, investigations have studied it in intact primary neurons. In this investigation, extracellular reduction of 2,6-dichloroindophenol (DCIP) and ferricyanide (FeCN) were measured as indicators of trans-plasma membrane electron transport by chick forebrain neurons. Neurons readily reduced DCIP, but not FeCN unless CoQ(1), an exogenous ubiquinone analog, was added to the assays. CoQ(1) stimulated FeCN reduction in a dose-dependent manner but had no effect on DCIP reduction. Reduction of both substrates was totally inhibited by epsilon-maleimidocaproic acid (MCA), a membrane-impermeant thiol reagent, and slightly inhibited by superoxide dismutase. Diphenylene iodonium, a flavoenzyme inhibitor, completely inhibited FeCN reduction but had no affect on DCIP reduction, suggesting that these substrates are reduced by distinct redox pathways. The relationship between plasma membrane electron transport and neuronal viability was tested using the inhibitors MCA and capsaicin. MCA caused a dose-dependent decline in neuronal viability that closely paralleled its inhibition of both reductase activities. Similarly capsaicin, a NADH oxidase inhibitor, induced a rapid decline in neuronal viability. These results suggest that trans-plasma membrane electron transport helps maintain a stable redox environment required for neuronal viability.
Collapse
Affiliation(s)
- M V Wright
- Institute of Arctic Biology, University of Alaska, Fairbanks, Alaska, USA
| | | |
Collapse
|
34
|
Morré DJ. Preferential inhibition of the plasma membrane NADH oxidase (NOX) activity by diphenyleneiodonium chloride with NADPH as donor. Antioxid Redox Signal 2002; 4:207-12. [PMID: 11970854 DOI: 10.1089/152308602753625960] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The cell-surface NADH oxidase (NOX) protein of plant and animal cells will utilize both NADH and NADPH as reduced electron donors for activity. The two activities are distinguished by a differential inhibition by the redox inhibitor diphenyleneiodonium chloride (DPI). Using both plasma membranes and cells, activity with NADPH as donor was markedly inhibited by DPI at submicromolar concentrations, whereas with NADH as donor, DPI was much less effective or had no effect on the activity. The possibility of the inhibition being the result of two different enzymes was eliminated by the use of a recombinant NOX protein. The findings support the concept that NOX proteins serve as terminal oxidases for plasma membrane electron transport involving cytosolic reduced pyridine nucleotides as the natural electron donors and with molecular oxygen as the electron acceptor.
Collapse
Affiliation(s)
- D James Morré
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907-1333, USA.
| |
Collapse
|
35
|
Berridge MV, Tan AS. High-capacity redox control at the plasma membrane of mammalian cells: trans-membrane, cell surface, and serum NADH-oxidases. Antioxid Redox Signal 2000; 2:231-42. [PMID: 11229528 DOI: 10.1089/ars.2000.2.2-231] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The high capacity of proliferating mammalian cells to transfer electrons from cytosolic NADH to extracellular electron acceptors like oxygen is poorly understood and not widely recognized. Nevertheless, trans-plasma membrane electron transport (plasma membrane redox control) probably ranks alongside the Na+/H+ antiport system (pH control) and glucose transport in facilitating cellular responses to physiological stimuli. These plasma membrane transport systems are acutely responsive to receptor ligation by growth factors, polypeptide hormones, and other cell activators. A novel tetrazolium-based cell proliferation assay that we have shown to measure an NADH-oxidoreductase component of the trans-plasma membrane electron transport system has allowed direct comparisons with NADH:ferricyanide-oxidoreductase and respiratory burst NADPH-oxidoreductase. In addition, an NAD(P)H-oxidase at the cell surface and an NADH-oxidase activity in body fluids can be measured by modifying the basic cell proliferation assay. As determined by reduction of the cell-impermeable tetrazolium reagent, WST-1, electron transfer across the plasma membrane of dividing cells can exceed that of fully activated human peripheral blood neutrophils. Cellular reduction of WST-1 is dependent on the presence of an intermediate electron acceptor and is inhibited by superoxide dismutase (SOD) and by oxygen, implying indirect involvement of superoxide in WST-1 reduction. Cell-surface NAD(P)H-oxidase and serum NADH-oxidase are shown to be distinct from trans-plasma membrane NADH-oxidoreductase by their differential sensitivity to capsaicin and pCMBS. The glycolytic metabolism of cancer cells may be linked to changes in trans-plasma membrane NADH:WST-1-oxidoreductase activity and to increased serum NADH-oxidase in cancer.
Collapse
Affiliation(s)
- M V Berridge
- Malaghan Institute of Medical Research, Wellington School of Medicine, Wellington South, New Zealand.
| | | |
Collapse
|