101
|
Tapia-Rojas C, Carvajal FJ, Mira RG, Arce C, Lerma-Cabrera JM, Orellana JA, Cerpa W, Quintanilla RA. Adolescent Binge Alcohol Exposure Affects the Brain Function Through Mitochondrial Impairment. Mol Neurobiol 2017; 55:4473-4491. [PMID: 28674997 DOI: 10.1007/s12035-017-0613-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/11/2017] [Indexed: 12/20/2022]
Abstract
In the young population, binge drinking is a pattern of problematic alcohol consumption, characterized by a short period of heavy drinking followed by abstinence which is frequently repeated over time. This drinking pattern is associated with mental problems, use of other drugs, and an increased risk of excessive alcohol intake during adulthood. However, little is known about the effects of binge drinking on brain function in adolescents and its neurobiological impact during the adulthood. In the present study, we evaluated the effects of alcohol on hippocampal memory, synaptic plasticity, and mitochondrial function in adolescent rats after a binge drinking episode in vivo. These effects were analyzed at 1, 3, or 7 weeks post alcohol exposure. Our results showed that binge-like ethanol pre-treated (BEP) rats exhibited early alterations in learning and memory tests accompanied by an impairment of synaptic plasticity that was total and partially compensated, respectively. These changes could be attributed to a rapid increase in oxidative damage and a late inflammatory response induced by post ethanol exposure. Additionally, BEP alters the regulation of mitochondrial dynamics and modifies the expression of mitochondrial permeability transition pore (mPTP) components, such as cyclophilin D (Cyp-D) and the voltage-dependent anion channel (VDAC). These mitochondrial structural changes result in the impairment of mitochondrial bioenergetics, decreasing ATP production progressively until adulthood. These results strongly suggest that teenage alcohol binge drinking impairs the function of the adult hippocampus including memory and synaptic plasticity as a consequence of the mitochondrial damage induced by alcohol and that the recovery of hippocampal function could implicate the activation of alternative pathways that fail to reestablish mitochondrial function.
Collapse
Affiliation(s)
- Cheril Tapia-Rojas
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
- Laboratory of Neurodegenerative Diseases, CIB, Universidad Autónoma de Chile, El llano Subercaseaux 2801, 5to Piso, San Miguel, 8910000, Santiago, Chile
| | - Francisco J Carvajal
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile
| | - Rodrigo G Mira
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile
| | - Camila Arce
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile
| | | | - Juan A Orellana
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Santiago, Chile
| | - Waldo Cerpa
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile.
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Rodrigo A Quintanilla
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile.
- Laboratory of Neurodegenerative Diseases, CIB, Universidad Autónoma de Chile, El llano Subercaseaux 2801, 5to Piso, San Miguel, 8910000, Santiago, Chile.
| |
Collapse
|
102
|
Prdx6 Upregulation by Curcumin Attenuates Ischemic Oxidative Damage via SP1 in Rats after Stroke. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6597401. [PMID: 28596967 PMCID: PMC5449737 DOI: 10.1155/2017/6597401] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/26/2017] [Indexed: 12/11/2022]
Abstract
Background The role of Peroxiredoxin 6 (Prdx6) in brain ischemia remains unclear. Curcumin (Cur) treatment elicits neuroprotective effects against cerebral ischemic injury, and the associated mechanisms may involve Prdx6. In this study, we investigated whether Prdx6 and the transcription factor specific protein 1 (SP1) were involved in the antioxidant effect of Cur after stoke. Methods Focal cerebral ischemic injury was induced by transient middle cerebral artery occlusion for 2 hours in male Sprague-Dawley rats treated with or without Prdx6 siRNA. Expression of Prdx6 in the penumbra was assessed by Real-Time PCR (RT-PCR), Western blot analysis, and immunoflourescent staining. In addition, infarct volume, neurological deficit score, and oxidative stress were evaluated. Prdx6 levels were also determined in the presence and absence of SP1 antagonist mithramycin A (MTM-A). Results Cur treatment upregulated Prdx6 protein expression and the number of Prdx6-positive neuronal cells 24 hours after reperfusion. Cur treatment also attenuated oxidative stress and induced neuroprotective effects against ischemic damage, whereas the beneficial effects of Cur treatment were lost in animals treated with Prdx6-siRNA. Prdx6 upregulation by Cur treatment was abolished by SP1 antagonists MTM. Conclusions Prdx6 upregulation by Cur treatment attenuates ischemic oxidative damage through SP1 induction in rats after stroke. This represents a novel mechanism of Cur-induced neuroprotection against cerebral ischemia.
Collapse
|
103
|
Debevec T, Millet GP, Pialoux V. Hypoxia-Induced Oxidative Stress Modulation with Physical Activity. Front Physiol 2017; 8:84. [PMID: 28243207 PMCID: PMC5303750 DOI: 10.3389/fphys.2017.00084] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/30/2017] [Indexed: 12/17/2022] Open
Abstract
Increased oxidative stress, defined as an imbalance between prooxidants and antioxidants, resulting in molecular damage and disruption of redox signaling, is associated with numerous pathophysiological processes and known to exacerbate chronic diseases. Prolonged systemic hypoxia, induced either by exposure to terrestrial altitude or a reduction in ambient O2 availability is known to elicit oxidative stress and thereby alter redox balance in healthy humans. The redox balance modulation is also highly dependent on the level of physical activity. For example, both high-intensity exercise and inactivity, representing the two ends of the physical activity spectrum, are known to promote oxidative stress. Numerous to-date studies indicate that hypoxia and exercise can exert additive influence upon redox balance alterations. However, recent evidence suggests that moderate physical activity can attenuate altitude/hypoxia-induced oxidative stress during long-term hypoxic exposure. The purpose of this review is to summarize recent findings on hypoxia-related oxidative stress modulation by different activity levels during prolonged hypoxic exposures and examine the potential mechanisms underlying the observed redox balance changes. The paper also explores the applicability of moderate activity as a strategy for attenuating hypoxia-related oxidative stress. Moreover, the potential of such moderate intensity activities used to counteract inactivity-related oxidative stress, often encountered in pathological, elderly and obese populations is also discussed. Finally, future research directions for investigating interactive effects of altitude/hypoxia and exercise on oxidative stress are proposed.
Collapse
Affiliation(s)
- Tadej Debevec
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan InstituteLjubljana, Slovenia
| | - Grégoire P. Millet
- Faculty of Biology and Medicine, Institute of Sport Sciences, University of LausanneLausanne, Switzerland
| | - Vincent Pialoux
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ Lyon, Université Claude Bernard Lyon 1Villeurbanne, France
- Institut Universitaire de FranceParis, France
| |
Collapse
|
104
|
Tubbs E, Rieusset J. Metabolic signaling functions of ER-mitochondria contact sites: role in metabolic diseases. J Mol Endocrinol 2017; 58:R87-R106. [PMID: 27965371 DOI: 10.1530/jme-16-0189] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 12/16/2022]
Abstract
Beyond the maintenance of cellular homeostasis and the determination of cell fate, ER-mitochondria contact sites, defined as mitochondria-associated membranes (MAM), start to emerge as an important signaling hub that integrates nutrient and hormonal stimuli and adapts cellular metabolism. Here, we summarize the established structural and functional features of MAM and mainly focus on the latest breakthroughs highlighting a crucial role of organelle crosstalk in the control of metabolic homeostasis. Lastly, we discuss recent studies that have revealed the importance of MAM in not only metabolic diseases but also in other pathologies with disrupted metabolism, shedding light on potential common molecular mechanisms and leading hopefully to novel treatment strategies.
Collapse
Affiliation(s)
- Emily Tubbs
- Department of Clinical SciencesLund University Diabetes Centre, Malmö, Sweden
| | - Jennifer Rieusset
- INSERM UMR-1060CarMeN Laboratory, Lyon 1 University, INRA U1235, INSA of Lyon, Charles Merieux Lyon-Sud medical Universities, Lyon, France
| |
Collapse
|
105
|
Duan Y, Sun F, Que S, Li Y, Yang S, Liu G. Prepregnancy maternal diabetes combined with obesity impairs placental mitochondrial function involving Nrf2/ARE pathway and detrimentally alters metabolism of offspring. Obes Res Clin Pract 2017; 12:90-100. [PMID: 28111084 DOI: 10.1016/j.orcp.2017.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/29/2016] [Accepted: 01/05/2017] [Indexed: 02/06/2023]
Abstract
Metabolic disorders usually increase the level of reactive oxygen species (ROS) and damage mitochondrial function. The placenta supplies nutrients and hormonal signals to the fetus for regulating fetal metabolism, and is also prone to injury by oxidants. The aim of this study was to determine the effect of pre-existing maternal type 2 diabetes mellitus (DM) combined with obesity on placental mitochondrial function and metabolism disorders of offspring. The study included 96 pregnant women. The women were put into the following groups: healthy women (control, n=24), women with DM (DM, n=24), women with obesity (OB, n=24) and women with both DM and obesity (DM+OB, n=24). The ROS level, mitochondrial content, and the mitochondrial respiratory complex activities of the placenta were measured in the four groups. The expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) was detected by immunofluorescence staining and western blotting. In addition, serum levels of insulin, glucose, leptin, nonesterified fatty acid (NEFA), adiponectin and triglycerides of their offspring were also measured. Maternal DM combined with obesity markedly increased ROS level, reduced mitochondrial DNA (mtDNA) content and mitochondrial respiratory complex I, II-III activities in placenta compared to the placenta from the control group and the DM group. Maternal DM combined with obesity significantly decreased Nrf2 and HO-1 expression. Furthermore, maternal DM combined with obesity influenced the glucose and lipid metabolism in their offspring. In conclusion, women with both DM and obesity detrimentally alter placenta function in oxidative stress regulation, and the Nrf2/ARE (antioxidant responsive element) pathway is involved. This may increase metabolic disturbance susceptibility in their offspring.
Collapse
Affiliation(s)
- Yang Duan
- Department of Neonatology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Fuqiang Sun
- Department of Neonatology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Shengshun Que
- Department of Neonatology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yueqin Li
- Department of Neonatology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Suyan Yang
- Department of Neonatology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Geli Liu
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300053, China.
| |
Collapse
|
106
|
Areti A, Yerra VG, Komirishetty P, Kumar A. Potential Therapeutic Benefits of Maintaining Mitochondrial Health in Peripheral Neuropathies. Curr Neuropharmacol 2017; 14:593-609. [PMID: 26818748 PMCID: PMC4981743 DOI: 10.2174/1570159x14666151126215358] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/31/2015] [Accepted: 11/24/2015] [Indexed: 02/07/2023] Open
Abstract
Background: Peripheral neuropathies are a group of diseases characterized by malfunctioning of peripheral nervous system. Neuropathic pain, one of the core manifestations of peripheral neuropathy remains as the most severe disabling condition affecting the social and daily routine life of patients suffering from peripheral neuropathy. Method: The current review is aimed at unfolding the possible role of mitochondrial dysfunction in peripheral nerve damage and to discuss on the probable therapeutic strategies against neuronal mitotoxicity. The article also highlights the therapeutic significance of maintaining a healthy mitochondrial environment in neuronal cells via pharmacological management in context of peripheral neuropathies. Results: Aberrant cellular signaling coupled with changes in neurotransmission, peripheral and central sensitization are found to be responsible for the pathogenesis of variant toxic neuropathies. Current research reports have indicated the possible involvement of mitochondria mediated redox imbalance as one of the principal causes of neuropathy aetiologies. In addition to imbalance in redox homeostasis, mitochondrial dysfunction is also responsible for alterations in physiological bioenergetic metabolism, apoptosis and autophagy pathways. Conclusions: In spite of various etiological factors, mitochondrial dysfunction has been found to be a major pathomechanism underlying the neuronal dysfunction associated with peripheral neuropathies. Pharmacological modulation of mitochondria either directly or indirectly is expected to yield therapeutic relief from various primary and secondary mitochondrial diseases.
Collapse
Affiliation(s)
| | | | | | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, Hyderabad, TG-500037.
| |
Collapse
|
107
|
Brahmbhatt A, Remuzzi A, Franzoni M, Misra S. The molecular mechanisms of hemodialysis vascular access failure. Kidney Int 2017; 89:303-316. [PMID: 26806833 PMCID: PMC4734360 DOI: 10.1016/j.kint.2015.12.019] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/20/2015] [Indexed: 01/01/2023]
Abstract
The arteriovenous fistula has been used for more than 50 years to provide vascular access for patients undergoing hemodialysis. More than 1.5 million patients worldwide have end stage renal disease and this population will continue to grow. The arteriovenous fistula is the preferred vascular access for patients, but its patency rate at 1 year is only 60%. The majority of arteriovenous fistulas fail because of intimal hyperplasia. In recent years, there have been many studies investigating the molecular mechanisms responsible for intimal hyperplasia and subsequent thrombosis. These studies have identified common pathways including inflammation, uremia, hypoxia, sheer stress, and increased thrombogenicity. These cellular mechanisms lead to increased proliferation, migration, and eventually stenosis. These pathways work synergistically through shared molecular messengers. In this review, we will examine the literature concerning the molecular basis of hemodialysis vascular access malfunction.
Collapse
Affiliation(s)
- Akshaar Brahmbhatt
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Remuzzi
- Biomedical Engineering Department, IRCCS—Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
- Engineering Department, University of Bergamo, Dalmine, Italy
| | - Marco Franzoni
- Biomedical Engineering Department, IRCCS—Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
108
|
Ribas V, Drew BG, Zhou Z, Phun J, Kalajian NY, Soleymani T, Daraei P, Widjaja K, Wanagat J, de Aguiar Vallim TQ, Fluitt AH, Bensinger S, Le T, Radu C, Whitelegge JP, Beaven SW, Tontonoz P, Lusis AJ, Parks BW, Vergnes L, Reue K, Singh H, Bopassa JC, Toro L, Stefani E, Watt MJ, Schenk S, Akerstrom T, Kelly M, Pedersen BK, Hewitt SC, Korach KS, Hevener AL. Skeletal muscle action of estrogen receptor α is critical for the maintenance of mitochondrial function and metabolic homeostasis in females. Sci Transl Med 2016; 8:334ra54. [PMID: 27075628 DOI: 10.1126/scitranslmed.aad3815] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/28/2016] [Indexed: 12/12/2022]
Abstract
Impaired estrogen receptor α (ERα) action promotes obesity and metabolic dysfunction in humans and mice; however, the mechanisms underlying these phenotypes remain unknown. Considering that skeletal muscle is a primary tissue responsible for glucose disposal and oxidative metabolism, we established that reduced ERα expression in muscle is associated with glucose intolerance and adiposity in women and female mice. To test this relationship, we generated muscle-specific ERα knockout (MERKO) mice. Impaired glucose homeostasis and increased adiposity were paralleled by diminished muscle oxidative metabolism and bioactive lipid accumulation in MERKO mice. Aberrant mitochondrial morphology, overproduction of reactive oxygen species, and impairment in basal and stress-induced mitochondrial fission dynamics, driven by imbalanced protein kinase A-regulator of calcineurin 1-calcineurin signaling through dynamin-related protein 1, tracked with reduced oxidative metabolism in MERKO muscle. Although muscle mitochondrial DNA (mtDNA) abundance was similar between the genotypes, ERα deficiency diminished mtDNA turnover by a balanced reduction in mtDNA replication and degradation. Our findings indicate the retention of dysfunctional mitochondria in MERKO muscle and implicate ERα in the preservation of mitochondrial health and insulin sensitivity as a defense against metabolic disease in women.
Collapse
Affiliation(s)
- Vicent Ribas
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Brian G Drew
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Zhenqi Zhou
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Jennifer Phun
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Nareg Y Kalajian
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Teo Soleymani
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Pedram Daraei
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Kevin Widjaja
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Jonathan Wanagat
- Division of Geriatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | - Amy H Fluitt
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA. Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Steven Bensinger
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Thuc Le
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA. Ahmanson Translational Imaging Division, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Caius Radu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA. Ahmanson Translational Imaging Division, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Julian P Whitelegge
- Pasarow Mass Spectrometry Laboratory and Neuropsychiatric Institute-Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Simon W Beaven
- Howard Hughes Medical Institute and Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Howard Hughes Medical Institute and Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Aldons J Lusis
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA. Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Brian W Parks
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Harpreet Singh
- Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Jean C Bopassa
- Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ligia Toro
- Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Enrico Stefani
- Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Matthew J Watt
- Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thorbjorn Akerstrom
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen 2200, Denmark
| | - Meghan Kelly
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen 2200, Denmark
| | - Bente K Pedersen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sylvia C Hewitt
- Receptor Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC 27709, USA
| | - Kenneth S Korach
- Receptor Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC 27709, USA
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA. UCLA Iris Cantor Women's Health Research Center, Los Angeles, CA 90095, USA.
| |
Collapse
|
109
|
Role of Mitochondria-Associated Endoplasmic Reticulum Membrane in Inflammation-Mediated Metabolic Diseases. Mediators Inflamm 2016; 2016:1851420. [PMID: 28074080 PMCID: PMC5198184 DOI: 10.1155/2016/1851420] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/17/2016] [Indexed: 12/11/2022] Open
Abstract
Inflammation is considered to be one of the most critical factors involved in the development of complex metabolic diseases such as type 2 diabetes, cancer, and cardiovascular disease. A few decades ago, the discovery of mitochondria-associated endoplasmic reticulum (ER) membrane (MAM) was followed by the identification of its roles in regulating cellular homeostatic processes, ranging from cellular bioenergetics to apoptosis. MAM provides an excellent platform for numerous signaling pathways; among them, inflammatory signaling pathways associated with MAM play a critical role in cellular defense during pathogenic infections and metabolic disorders. However, induction of MAM causes deleterious effects by amplifying mitochondrial reactive oxygen species generation through increased calcium transfer from the ER to mitochondria, thereby causing mitochondrial damage and release of mitochondrial components into the cytosol as damage-associated molecular patterns (DAMPs). These mitochondrial DAMPs rapidly activate MAM-resident inflammasome components and other inflammatory factors, which promote inflammasome complex formation and release of proinflammatory cytokines in pathological conditions. Long-term stimulation of the inflammasome instigates chronic inflammation, leading to the pathogenesis of metabolic diseases. In this review, we summarize the current understanding of MAM and its association with inflammation-mediated metabolic diseases.
Collapse
|
110
|
Zhang Y, Ji X, Ku T, Sang N. Inflammatory response and endothelial dysfunction in the hearts of mice co-exposed to SO 2 , NO 2 , and PM 2.5. ENVIRONMENTAL TOXICOLOGY 2016; 31:1996-2005. [PMID: 26417707 DOI: 10.1002/tox.22200] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 09/12/2015] [Accepted: 09/13/2015] [Indexed: 06/05/2023]
Abstract
SO2 , NO2 , and PM2.5 are typical air pollutants produced during the combustion of coal. Increasing evidence indicates that air pollution has contributed to the development and progression of heart-related diseases over the past decades. However, little experimental data and few studies of SO2 , NO2 , and PM2.5 co-exposure in animals exist; therefore, the relevant mechanisms underlying this phenomenon are unclear. An important characteristic of air pollution is that co-exposure persists at a low concentration throughout a lifetime. In the present study, we treated adult mice with SO2 , NO2 , and PM2.5 at various concentrations (0.5 mg/m3 SO2 , 0.2 mg/m3 NO2 6 h/d, with intranasal instillation of 1 mg/kg PM2.5 every other day during these exposures; or 3.5 mg/m3 SO2 , 2 mg/m3 NO2 6 h/d, and 10 mg/kg PM2.5 for 28 d). Blood pressure (BP), heart rate (HR), histopathological damage, and inflammatory and endothelial cytokines in the heart were assessed. The results indicate that co-exposure caused endothelial dysfunction by elevating endothelin-1 (ET-1) expression and repressing the endothelial nitric oxide synthase (eNOS) level as well as stimulating the inflammatory response by increasing the levels of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Additionally, these alterations were confirmed by histological staining. Furthermore, we observed decreased BP and increased HR after co-exposure. Our results indicate that co-exposure to SO2 , NO2 , and PM2.5 may be a major risk factor for cardiac disease and may induce injury to the hearts of mammals and contribute to heart disease. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1996-2005, 2016.
Collapse
Affiliation(s)
- Yingying Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, People's Republic of China
| |
Collapse
|
111
|
Zakharova VV, Pletjushkina OY, Zinovkin RA, Popova EN, Chernyak BV. Mitochondria-Targeted Antioxidants and Uncouplers of Oxidative Phosphorylation in Treatment of the Systemic Inflammatory Response Syndrome (SIRS). J Cell Physiol 2016; 232:904-912. [DOI: 10.1002/jcp.25626] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Vlada V. Zakharova
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
- Faculty of Bioengineering and Bioinformatics; Lomonosov Moscow State University; Moscow Russia
| | - Olga Yu. Pletjushkina
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| | - Roman A. Zinovkin
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| | - Ekaterina N. Popova
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| | - Boris V. Chernyak
- Belozersky Institute of Physico-Chemical Biology; Lomonosov Moscow State University; Moscow Russia
| |
Collapse
|
112
|
Caligiuri A, Gentilini A, Marra F. Molecular Pathogenesis of NASH. Int J Mol Sci 2016; 17:ijms17091575. [PMID: 27657051 PMCID: PMC5037841 DOI: 10.3390/ijms17091575] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the main cause of chronic liver disease in the Western world and a major health problem, owing to its close association with obesity, diabetes, and the metabolic syndrome. NASH progression results from numerous events originating within the liver, as well as from signals derived from the adipose tissue and the gastrointestinal tract. In a fraction of NASH patients, disease may progress, eventually leading to advanced fibrosis, cirrhosis and hepatocellular carcinoma. Understanding the mechanisms leading to NASH and its evolution to cirrhosis is critical to identifying effective approaches for the treatment of this condition. In this review, we focus on some of the most recent data reported on the pathogenesis of NASH and its fibrogenic progression, highlighting potential targets for treatment or identification of biomarkers of disease progression.
Collapse
Affiliation(s)
- Alessandra Caligiuri
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Alessandra Gentilini
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| |
Collapse
|
113
|
Boeck C, Koenig AM, Schury K, Geiger ML, Karabatsiakis A, Wilker S, Waller C, Gündel H, Fegert JM, Calzia E, Kolassa IT. Inflammation in adult women with a history of child maltreatment: The involvement of mitochondrial alterations and oxidative stress. Mitochondrion 2016; 30:197-207. [DOI: 10.1016/j.mito.2016.08.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/15/2016] [Accepted: 08/12/2016] [Indexed: 12/24/2022]
|
114
|
Eike LM, Yang N, Rekdal Ø, Sveinbjørnsson B. The oncolytic peptide LTX-315 induces cell death and DAMP release by mitochondria distortion in human melanoma cells. Oncotarget 2016; 6:34910-23. [PMID: 26472184 PMCID: PMC4741498 DOI: 10.18632/oncotarget.5308] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/01/2015] [Indexed: 02/01/2023] Open
Abstract
Host defense peptides (HDPs) are naturally occurring molecules found in most species, in which they play a significant role in the first line defense against intruding pathogens, and several HDPs have been shown to possess anticancer activity. Structure-activity relationship studies on the HDP bovine lactoferricin revealed a de novo design of a nonamer peptide LTX-315, with oncolytic properties. In the present study, we investigated the oncolytic activity of LTX-315 in human melanoma cells (A375). LTX-315 induced a rapid plasma membrane disruption and cell death within 2 hours. At a low concentration, fluorescence-labeled LTX-315 was internalized and accumulated in cytoplasmic vacuoles in close proximity to the mitochondria. The mitochondrial membrane potential was shown to depolarize as a consequence of LTX-315 treatment and at ultrastructural level, the mitochondria morphology was significantly altered. Release of danger signals (DAMPs) such as ATP, Cytochrome C and HMGB1 into the cell supernatant of cultured cells was evident minutes after peptide treatment. The oncolytic effect of LTX-315 involving perturbation of both the cell membrane and the mitochondria with subsequent release of DAMPs may highlight the ability of LTX-315 to induce complete regression and long-term protective immune responses as previously reported in experimental animal models.
Collapse
Affiliation(s)
- Liv-Marie Eike
- Department of Molecular Inflammation Research and Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Nannan Yang
- Department of Community Medicine, Faculty of Health University of Tromsø, Tromsø, Norway
| | - Øystein Rekdal
- Department of Molecular Inflammation Research and Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.,Lytix Biopharma, Oslo, Norway
| | - Baldur Sveinbjørnsson
- Department of Molecular Inflammation Research and Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| |
Collapse
|
115
|
Rimessi A, Previati M, Nigro F, Wieckowski MR, Pinton P. Mitochondrial reactive oxygen species and inflammation: Molecular mechanisms, diseases and promising therapies. Int J Biochem Cell Biol 2016; 81:281-293. [PMID: 27373679 DOI: 10.1016/j.biocel.2016.06.015] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/20/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
Over the last few decades, many different groups have been engaged in studies of new roles for mitochondria, particularly the coupling of alterations in the redox pathway with the inflammatory responses involved in different diseases, including Alzheimer's disease, Parkinson's disease, atherosclerosis, cerebral cavernous malformations, cystic fibrosis and cancer. Mitochondrial dysfunction is important in these pathological conditions, suggesting a pivotal role for mitochondria in the coordination of pro-inflammatory signaling from the cytosol and signaling from other subcellular organelles. In this regard, mitochondrial reactive oxygen species are emerging as perfect liaisons that can trigger the assembly and successive activation of large caspase-1- activating complexes known as inflammasomes. This review offers a glimpse into the mechanisms by which inflammasomes are activated by mitochondrial mechanisms, including reactive oxygen species production and mitochondrial Ca2+ uptake, and the roles they can play in several inflammatory pathologies.
Collapse
Affiliation(s)
- Alessandro Rimessi
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Maurizio Previati
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Human Anatomy and Histology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Federica Nigro
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Mariusz R Wieckowski
- Dept. of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland.
| | - Paolo Pinton
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
116
|
Li W, Jin D, Hata M, Takai S, Yamanishi K, Shen W, El-Darawish Y, Yamanishi H, Okamura H. Dysfunction of mitochondria and deformed gap junctions in the heart of IL-18-deficient mice. Am J Physiol Heart Circ Physiol 2016; 311:H313-25. [PMID: 27288439 DOI: 10.1152/ajpheart.00927.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 06/03/2016] [Indexed: 01/01/2023]
Abstract
Interleukin-18 (IL-18) was discovered as an interferon-γ-inducing factor and has been regarded as a proinflammatory cytokine. However, IL-18 is ubiquitously expressed both in immune/inflammatory cells and in nonimmune cells, and its biological roles have not been sufficiently elucidated. Here, we demonstrate that IL-18-deficient [IL-18 knockout (KO)] mice have heart abnormalities that may be related to impaired autophagy. In endurance running tests, IL-18KO mice ran significantly shorter distances compared with wild-type (WT) mice. Echocardiographs indicated disability in the systolic and diastolic functions of the IL-18KO mouse heart. Immunostaining of connexin 43 showed heterogeneous localization of gap junctions in the lateral membranes of the IL-18KO cardiac myocytes. Western blotting analysis revealed decreased phosphorylated connexin 43 in the IL-18KO heart. Electron microscopy revealed unusual localization of intercalated disks, swollen or damaged mitochondria, and broad, indistinct Z-lines in the IL-18KO heart. In accordance with the morphological observation, mitochondrial respiratory function, including that of complexes I and IV, was impaired, and production of reactive oxygen species was augmented in IL-18KO hearts. Notably, levels of LC3-II were markedly lower in the IL-18KO hearts than in WT hearts. In the culture of cardiac myocytes of IL-18KO neonates, exogenous IL-18 upregulated LC3-II and increased the number of intact mitochondria with high mitochondrial membrane potential. These results indicated that IL-18 has roles apart from those as a proinflammatory cytokine in cardiac myocytes and suggested that IL-18 contributes to the homeostatic maintenance of mitochondrial function and gap-junction turnover in cardiac myocytes, possibly by upregulating autophagy.
Collapse
Affiliation(s)
- Wen Li
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Hyogo, Japan
| | - Denan Jin
- Department of Pharmacology, Osaka Medical College, Osaka, Japan
| | - Masaki Hata
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Hyogo, Japan
| | - Shinji Takai
- Department of Pharmacology, Osaka Medical College, Osaka, Japan
| | - Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo College of Medicine, Hyogo, Japan
| | - Weili Shen
- Shanghai Key Laboratory of Hypertension, Shanghai, China; and
| | - Yosif El-Darawish
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Hyogo, Japan
| | | | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Hyogo, Japan;
| |
Collapse
|
117
|
Rosenberger DS, Falangola MF, Ledreux A, Nie X, Suhre WM, Boger HA, Granholm AC. Memory and hippocampal architecture following short-term midazolam in western diet-treated rats. Neurosci Lett 2016; 621:68-74. [PMID: 27080429 PMCID: PMC4853265 DOI: 10.1016/j.neulet.2016.04.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 03/15/2016] [Accepted: 04/09/2016] [Indexed: 01/01/2023]
Abstract
The impact of short-term benzodiazepine exposure on cognition in middle-aged or older patients is a highly debated topic among anesthesiologists, critical care physicians and public media. "Western diet" (WD) consumption is linked to impaired cognition as well. The combination of benzodiazepines with substantial exposure to WD might set the stage for increased hippocampal vulnerability for benzodiazepines leading to exaggerated cognitive impairment in the postoperative period. In this study, Fischer 344 rats were fed either WD or standard rodent diet from 5 to 10.5 months of age. Rats were exposed to midazolam or placebo two days prior to an MRI scan using Diffusional Kurtosis Imaging (DKI) to assess brain microstructural integrity, followed by behavioral testing using a water radial arm maze. Hippocampal tissue was collected to assess alterations in protein biochemistry in brain regions associated with learning and memory. Our results showed that rats exposed to the combination of midazolam and WD had significantly delayed time of learning and exhibited spatial memory impairment. Further, we observed an overall increase of kurtosis metrics in the hippocampus and increased expression of the mitochondrial protein VDAC2 in midazolam-treated rats. Our data suggest that both the short-acting benzodiazepine midazolam and WD contribute to negatively affect the brain in middle-aged rats. This study is the first application of DKI on the effects of midazolam and WD exposure, and the findings demonstrate that diffusion metrics are sensitive indicators of changes in the complexity of neurite architecture.
Collapse
Affiliation(s)
- Dorothea S Rosenberger
- Department of Anesthesiology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States.
| | - Maria F Falangola
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC 29425, United States; Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Aurélie Ledreux
- Department of Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Xingju Nie
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC 29425, United States; Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Wendy M Suhre
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, United States
| | - Heather A Boger
- Department of Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Ann-Charlotte Granholm
- Department of Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, United States; Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States
| |
Collapse
|
118
|
Mitochondrial H2O2 in Lung Antigen-Presenting Cells Blocks NF-κB Activation to Prevent Unwarranted Immune Activation. Cell Rep 2016; 15:1700-14. [PMID: 27184852 DOI: 10.1016/j.celrep.2016.04.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/10/2016] [Accepted: 04/15/2016] [Indexed: 12/21/2022] Open
Abstract
Inhalation of environmental antigens such as allergens does not always induce inflammation in the respiratory tract. While antigen-presenting cells (APCs), including dendritic cells and macrophages, take up inhaled antigens, the cell-intrinsic molecular mechanisms that prevent an inflammatory response during this process, such as activation of the transcription factor NF-κB, are not well understood. Here, we show that the nuclear receptor PPARγ plays a critical role in blocking NF-κB activation in response to inhaled antigens to preserve immune tolerance. Tolerance induction promoted mitochondrial respiration, generation of H2O2, and suppression of NF-κB activation in WT, but not PPARγ-deficient, APCs. Forced restoration of H2O2 in PPARγ-deficient cells suppressed IκBα degradation and NF-κB activation. Conversely, scavenging reactive oxygen species from mitochondria promoted IκBα degradation with loss of regulatory and promotion of inflammatory T cell responses in vivo. Thus, communication between PPARγ and the mitochondria maintains immune quiescence in the airways.
Collapse
|
119
|
Fanconi Anemia Proteins Function in Mitophagy and Immunity. Cell 2016; 165:867-81. [PMID: 27133164 DOI: 10.1016/j.cell.2016.04.006] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/29/2016] [Accepted: 03/24/2016] [Indexed: 01/09/2023]
Abstract
Fanconi anemia (FA) pathway genes are important tumor suppressors whose best-characterized function is repair of damaged nuclear DNA. Here, we describe an essential role for FA genes in two forms of selective autophagy. Genetic deletion of Fancc blocks the autophagic clearance of viruses (virophagy) and increases susceptibility to lethal viral encephalitis. Fanconi anemia complementation group C (FANCC) protein interacts with Parkin, is required in vitro and in vivo for clearance of damaged mitochondria, and decreases mitochondrial reactive oxygen species (ROS) production and inflammasome activation. The mitophagy function of FANCC is genetically distinct from its role in genomic DNA damage repair. Moreover, additional genes in the FA pathway, including FANCA, FANCF, FANCL, FANCD2, BRCA1, and BRCA2, are required for mitophagy. Thus, members of the FA pathway represent a previously undescribed class of selective autophagy genes that function in immunity and organellar homeostasis. These findings have implications for understanding the pathogenesis of FA and cancers associated with mutations in FA genes.
Collapse
|
120
|
Vartak-Sharma N, Nooka S, Ghorpade A. Astrocyte elevated gene-1 (AEG-1) and the A(E)Ging HIV/AIDS-HAND. Prog Neurobiol 2016; 157:133-157. [PMID: 27090750 DOI: 10.1016/j.pneurobio.2016.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/11/2016] [Accepted: 03/19/2016] [Indexed: 12/23/2022]
Abstract
Recent attempts to analyze human immunodeficiency virus (HIV)-1-induced gene expression changes in astrocytes uncovered a multifunctional oncogene, astrocyte elevated gene-1 (AEG-1). Our previous studies revealed that AEG-1 regulates reactive astrocytes proliferation, migration and inflammation, hallmarks of aging and CNS injury. Moreover, the involvement of AEG-1 in neurodegenerative disorders, such as Huntington's disease and migraine, and its induction in the aged brain suggest a plausible role in regulating overall CNS homeostasis and aging. Therefore, it is important to investigate AEG-1 specifically in aging-associated cognitive decline. In this study, we decipher the common mechanistic links in cancer, aging and HIV-1-associated neurocognitive disorders that likely contribute to AEG-1-based regulation of astrocyte responses and function. Despite AEG-1 incorporation into HIV-1 virions and its induction by HIV-1, tumor necrosis factor-α and interleukin-1β, the specific role(s) of AEG-1 in astrocyte-driven HIV-1 neuropathogenesis are incompletely defined. We propose that AEG-1 plays a central role in a multitude of cellular stress responses involving mitochondria, endoplasmic reticulum and the nucleolus. It is thus important to further investigate AEG-1-based cellular and molecular regulation in order to successfully develop better therapeutic approaches that target AEG-1 to combat cancer, HIV-1 and aging.
Collapse
Affiliation(s)
- Neha Vartak-Sharma
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107-2699, USA; Institute for Integrated Cell-Material Sciences, Kyoto University, Japan; Institute for Stem Cell Research and Regenerative Medicine, National Center for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Shruthi Nooka
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107-2699, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107-2699, USA.
| |
Collapse
|
121
|
The oxindole Syk inhibitor OXSI-2 blocks nigericin-induced inflammasome signaling and pyroptosis independent of potassium efflux. Biochem Biophys Res Commun 2016; 472:545-50. [DOI: 10.1016/j.bbrc.2016.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/06/2016] [Indexed: 12/30/2022]
|
122
|
Volt H, García JA, Doerrier C, Díaz-Casado ME, Guerra-Librero A, López LC, Escames G, Tresguerres JA, Acuña-Castroviejo D. Same molecule but different expression: aging and sepsis trigger NLRP3 inflammasome activation, a target of melatonin. J Pineal Res 2016; 60:193-205. [PMID: 26681113 DOI: 10.1111/jpi.12303] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/11/2015] [Indexed: 12/13/2022]
Abstract
The connection between the innate immune system, clock genes, and mitochondrial bioenergetics was analyzed during aging and sepsis in mouse heart. Our results suggest that the sole NF-κB activation does not explain the inflammatory process underlying aging; the former also triggers the NLRP3 inflammasome that enhances caspase-1-dependent maturation of IL-1β. In this way, aged mice enter into a vicious cycle as IL-1β further activates the NF-κB/NLRP3 inflammasome link. The origin of NF-κB activation was related to the age-dependent Bmal1/Clock/RORα/Rev-Erbα loop disruption, which lowers NAD(+) levels, reducing the SIRT1 deacetylase ability to inactivate NF-κB. Consequently, NF-κB binding to DNA increases, raising the formation of proinflammatory mediators and inducing mitochondrial impairment. The cycle is then closed with the subsequent NLRP3 inflammasome activation. This paired contribution of the innate immune pathways serves as a catalyst to magnify the response to sepsis in aged compared with young mice. Melatonin administration blunted the septic response, reducing inflammation and oxidative stress, and enhancing mitochondrial function at the levels of nonseptic aged mice, but it did not counteract the age-related inflammation. Together, our results suggest that, although with different strengths, chronoinflammaging constitutes the biochemical substrate of aging and sepsis, and identifies the NLRP3 inflammasome as a new molecular target for melatonin, providing a rationale for its use in NLRP3-dependent diseases.
Collapse
Affiliation(s)
- Huayqui Volt
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - José A García
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Carolina Doerrier
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - María E Díaz-Casado
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Ana Guerra-Librero
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Luis C López
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Germaine Escames
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Jesús A Tresguerres
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Darío Acuña-Castroviejo
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
123
|
cJun N-terminal kinase (JNK) phosphorylation of serine 36 is critical for p66Shc activation. Sci Rep 2016; 6:20930. [PMID: 26868434 PMCID: PMC4751440 DOI: 10.1038/srep20930] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/13/2016] [Indexed: 12/19/2022] Open
Abstract
p66Shc-dependent ROS production contributes to many pathologies including ischemia/reperfusion injury (IRI) during solid organ transplantation. Inhibiting p66Shc activation may provide a novel therapeutic approach to prevent damage, which is poorly managed by antioxidants in vivo. Previous work suggested that pro-oxidant and a pro-apoptotic function of p66Shc required mitochondrial import, which depended on serine 36 phosphorylation. PKCß has been proposed as S36 kinase but cJun N-terminal kinases (JNKs) may also phosphorylate this residue. To simulate the early stages of ischemia/reperfusion (IR) we either used H2O2 treatment or hypoxia/reoxygenation (HR). As during reperfusion in vivo, we observed increased JNK and p38 activity in mouse embryonic fibroblasts (MEFs) and HL-1 cardiomyocytes along with significantly increased p66ShcS36 phosphorylation, ROS production and cell damage. Application of specific inhibitors caused a pronounced decrease in p66ShcS36 phosphorylation only in the case of JNK1/2. Moreover, S36 phosphorylation of recombinant p66Shc by JNK1 but not PKCß was demonstrated. We further confirmed JNK1/2-dependent regulation of p66ShcS36 phosphorylation, ROS production and cell death using JNK1/2 deficient MEFs. Finally, the low ROS phenotype of JNK1/2 knockout MEFs was reversed by the phosphomimetic p66ShcS36E mutant. Inhibiting JNK1/2-regulated p66Shc activation may thus provide a therapeutic approach for the prevention of oxidative damage.
Collapse
|
124
|
Yoo SJ, Go E, Kim YE, Lee S, Kwon J. Roles of Reactive Oxygen Species in Rheumatoid Arthritis Pathogenesis. JOURNAL OF RHEUMATIC DISEASES 2016. [DOI: 10.4078/jrd.2016.23.6.340] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Su-Jin Yoo
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eunbyeol Go
- Department of Medical Education, Chungnam National University School of Medicine, Daejeon, Korea
| | - Ye-Eun Kim
- Department of Medical Education, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sunyoung Lee
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jaeyul Kwon
- Department of Medical Education, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
125
|
Abstract
In addition to oxidative phosphorylation (OXPHOS), mitochondria perform other functions such as heme biosynthesis and oxygen sensing and mediate calcium homeostasis, cell growth, and cell death. They participate in cell communication and regulation of inflammation and are important considerations in aging, drug toxicity, and pathogenesis. The cell's capacity to maintain its mitochondria involves intramitochondrial processes, such as heme and protein turnover, and those involving entire organelles, such as fusion, fission, selective mitochondrial macroautophagy (mitophagy), and mitochondrial biogenesis. The integration of these processes exemplifies mitochondrial quality control (QC), which is also important in cellular disorders ranging from primary mitochondrial genetic diseases to those that involve mitochondria secondarily, such as neurodegenerative, cardiovascular, inflammatory, and metabolic syndromes. Consequently, mitochondrial biology represents a potentially useful, but relatively unexploited area of therapeutic innovation. In patients with genetic OXPHOS disorders, the largest group of inborn errors of metabolism, effective therapies, apart from symptomatic and nutritional measures, are largely lacking. Moreover, the genetic and biochemical heterogeneity of these states is remarkably similar to those of certain acquired diseases characterized by metabolic and oxidative stress and displaying wide variability. This biologic variability reflects cell-specific and repair processes that complicate rational pharmacological approaches to both primary and secondary mitochondrial disorders. However, emerging concepts of mitochondrial turnover and dynamics along with new mitochondrial disease models are providing opportunities to develop and evaluate mitochondrial QC-based therapies. The goals of such therapies extend beyond amelioration of energy insufficiency and tissue loss and entail cell repair, cell replacement, and the prevention of fibrosis. This review summarizes current concepts of mitochondria as disease elements and outlines novel strategies to address mitochondrial dysfunction through the stimulation of mitochondrial biogenesis and quality control.
Collapse
Affiliation(s)
- Hagir B Suliman
- Departments of Medicine (C.A.P.), Anesthesiology (H.B.S.), Duke Cancer Institute (H.B.S.), and Pathology (C.A.P.), Duke University Medical Center, Durham North Carolina
| | - Claude A Piantadosi
- Departments of Medicine (C.A.P.), Anesthesiology (H.B.S.), Duke Cancer Institute (H.B.S.), and Pathology (C.A.P.), Duke University Medical Center, Durham North Carolina
| |
Collapse
|
126
|
Naquet P, Giessner C, Galland F. Metabolic adaptation of tissues to stress releases metabolites influencing innate immunity. Curr Opin Immunol 2015; 38:30-8. [PMID: 26605965 DOI: 10.1016/j.coi.2015.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/03/2015] [Accepted: 10/21/2015] [Indexed: 12/11/2022]
Abstract
Recent developments have demonstrated that metabolic rewiring imposed by adaptation of tissues to stress leads to the release of various metabolites which directly or indirectly impact innate immune responses and inflammation. Some metabolites can behave as second messengers and leave local cues in tissues. Immune cells which infiltrate stressed tissues reorient their metabolism to cope with these microenvironmental cues while preserving their effector functions in tissues.
Collapse
Affiliation(s)
- Philippe Naquet
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France.
| | - Caroline Giessner
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| | - Franck Galland
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| |
Collapse
|
127
|
Yoon CM, Nam M, Oh YM, Dela Cruz CS, Kang MJ. Mitochondrial Regulation of Inflammasome Activation in Chronic Obstructive Pulmonary Disease. J Innate Immun 2015; 8:121-8. [PMID: 26536345 DOI: 10.1159/000441299] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 09/25/2015] [Indexed: 12/14/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by enhanced chronic airway and lung inflammatory responses to noxious particles or gases. It is a major unmet medical need worldwide, and in Western society is strongly associated with exposure to cigarette smoke (CS). CS-induced inflammation is believed to be a key immune driver in the pathogenesis of COPD. Since the concept of inflammasomes was first introduced nearly a decade ago, these have been increasingly recognized as a central player in innate immune and inflammatory responses. In addition, studies have emerged demonstrating that mitochondrial innate immune signaling plays an important role in CS-induced inflammasome activation, pulmonary inflammation and tissue remodeling responses. Here, recent discoveries about inflammasome activation and mitochondrial biology and their role in COPD pathogenesis are reviewed. In addition, the current limitations of our understanding of this theme and future research directions are discussed.
Collapse
Affiliation(s)
- Chang Min Yoon
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Conn., USA
| | | | | | | | | |
Collapse
|
128
|
Yaron JR, Gangaraju S, Rao MY, Kong X, Zhang L, Su F, Tian Y, Glenn HL, Meldrum DR. K(+) regulates Ca(2+) to drive inflammasome signaling: dynamic visualization of ion flux in live cells. Cell Death Dis 2015; 6:e1954. [PMID: 26512962 PMCID: PMC5399176 DOI: 10.1038/cddis.2015.277] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/27/2015] [Accepted: 08/31/2015] [Indexed: 01/05/2023]
Abstract
P2X7 purinergic receptor engagement with extracellular ATP induces transmembrane potassium and calcium flux resulting in assembly of the NLRP3 inflammasome in LPS-primed macrophages. The role of potassium and calcium in inflammasome regulation is not well understood, largely due to limitations in existing methods for interrogating potassium in real time. The use of KS6, a novel sensor for selective and sensitive dynamic visualization of intracellular potassium flux in live cells, multiplexed with the intracellular calcium sensor Fluo-4, revealed a coordinated relationship between potassium and calcium. Interestingly, the mitochondrial potassium pool was mobilized in a P2X7 signaling, and ATP dose-dependent manner, suggesting a role for mitochondrial sensing of cytosolic ion perturbation. Through treatment with extracellular potassium we found that potassium efflux was necessary to permit sustained calcium entry, but not transient calcium flux from intracellular stores. Further, intracellular calcium chelation with BAPTA-AM indicated that P2X7-induced potassium depletion was independent of calcium mobilization. This evidence suggests that both potassium efflux and calcium influx are necessary for mitochondrial reactive oxygen generation upstream of NLRP3 inflammasome assembly and pyroptotic cell death. We propose a model wherein potassium efflux is necessary for calcium influx, resulting in mitochondrial reactive oxygen generation to trigger the NLRP3 inflammasome.
Collapse
Affiliation(s)
- J R Yaron
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA.,Biological Design Graduate Program, School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, 85287 AZ, USA
| | - S Gangaraju
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| | - M Y Rao
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| | - X Kong
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| | - L Zhang
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| | - F Su
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| | - Y Tian
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| | - H L Glenn
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| | - D R Meldrum
- Center for Biosignatures Discovery Automation, The Biodesign Institute, Arizona State University, Tempe, 85287 AZ, USA
| |
Collapse
|
129
|
Riquelme SA, Pogu J, Anegon I, Bueno SM, Kalergis AM. Carbon monoxide impairs mitochondria-dependent endosomal maturation and antigen presentation in dendritic cells. Eur J Immunol 2015; 45:3269-88. [DOI: 10.1002/eji.201545671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 08/18/2015] [Accepted: 09/24/2015] [Indexed: 01/07/2023]
Affiliation(s)
- Sebastián A. Riquelme
- Millennium Institute on Immunology and Immunotherapy; Departamento de Genética Molecular y Microbiología; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile; Santiago Chile
- INSERM; UMR 1064; CHU Nantes; ITUN; Université de Nantes; Faculté de Médecine; Nantes France
| | - Julien Pogu
- INSERM; UMR 1064; CHU Nantes; ITUN; Université de Nantes; Faculté de Médecine; Nantes France
| | - Ignacio Anegon
- INSERM; UMR 1064; CHU Nantes; ITUN; Université de Nantes; Faculté de Médecine; Nantes France
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy; Departamento de Genética Molecular y Microbiología; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile; Santiago Chile
- INSERM; UMR 1064; CHU Nantes; ITUN; Université de Nantes; Faculté de Médecine; Nantes France
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy; Departamento de Genética Molecular y Microbiología; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile; Santiago Chile
- INSERM; UMR 1064; CHU Nantes; ITUN; Université de Nantes; Faculté de Médecine; Nantes France
- Departamento de Inmunología Clínica y Reumatología; Facultad de Medicina, Pontificia Universidad Católica de Chile; Santiago Chile
| |
Collapse
|
130
|
Mitochondrial ROS Induces Cardiac Inflammation via a Pathway through mtDNA Damage in a Pneumonia-Related Sepsis Model. PLoS One 2015; 10:e0139416. [PMID: 26448624 PMCID: PMC4598156 DOI: 10.1371/journal.pone.0139416] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/14/2015] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that mitochondria-targeted vitamin E (Mito-Vit-E), a mtROS specific antioxidant, improves cardiac performance and attenuates inflammation in a pneumonia-related sepsis model. In this study, we applied the same approaches to decipher the signaling pathway(s) of mtROS-dependent cardiac inflammation after sepsis. Sepsis was induced in Sprague Dawley rats by intratracheal injection of S. pneumoniae. Mito-Vit-E, vitamin E or vehicle was administered 30 minutes later. In myocardium 24 hours post-inoculation, Mito-Vit-E, but not vitamin E, significantly protected mtDNA integrity and decreased mtDNA damage. Mito-Vit-E alleviated sepsis-induced reduction in mitochondria-localized DNA repair enzymes including DNA polymerase γ, AP endonuclease, 8-oxoguanine glycosylase, and uracil-DNA glycosylase. Mito-Vit-E dramatically improved metabolism and membrane integrity in mitochondria, suppressed leakage of mtDNA into the cytoplasm, inhibited up-regulation of Toll-like receptor 9 (TLR9) pathway factors MYD88 and RAGE, and limited RAGE interaction with its ligand TFAM in septic hearts. Mito-Vit-E also deactivated NF-κB and caspase 1, reduced expression of the essential inflammasome component ASC, and decreased inflammatory cytokine IL–1β. In vitro, both Mito-Vit-E and TLR9 inhibitor OND-I suppressed LPS-induced up-regulation in MYD88, RAGE, ASC, active caspase 1, and IL–1β in cardiomyocytes. Since free mtDNA escaped from damaged mitochondria function as a type of DAMPs to stimulate inflammation through TLR9, these data together suggest that sepsis-induced cardiac inflammation is mediated, at least partially, through mtDNA-TLR9-RAGE. At last, Mito-Vit-E reduced the circulation of myocardial injury marker troponin-I, diminished apoptosis and amended morphology in septic hearts, suggesting that mitochondria-targeted antioxidants are a potential cardioprotective approach for sepsis.
Collapse
|
131
|
Novak EA, Mollen KP. Mitochondrial dysfunction in inflammatory bowel disease. Front Cell Dev Biol 2015; 3:62. [PMID: 26484345 PMCID: PMC4589667 DOI: 10.3389/fcell.2015.00062] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 09/14/2015] [Indexed: 12/12/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) represents a group of idiopathic disorders characterized by chronic or recurring inflammation of the gastrointestinal tract. While the exact etiology of disease is unknown, IBD is recognized to be a complex, multifactorial disease that results from an intricate interplay of genetic predisposition, an altered immune response, changes in the intestinal microbiota, and environmental factors. Together, these contribute to a destruction of the intestinal epithelial barrier, increased gut permeability, and an influx of immune cells. Given that most cellular functions as well as maintenance of the epithelial barrier is energy-dependent, it is logical to assume that mitochondrial dysfunction may play a key role in both the onset and recurrence of disease. Indeed several studies have demonstrated evidence of mitochondrial stress and alterations in mitochondrial function within the intestinal epithelium of patients with IBD and mice undergoing experimental colitis. Although the hallmarks of mitochondrial dysfunction, including oxidative stress and impaired ATP production are known to be evident in the intestines of patients with IBD, it is as yet unclear whether these processes occur as a cause of consequence of disease. We provide a current review of mitochondrial function in the setting of intestinal inflammation during IBD.
Collapse
Affiliation(s)
- Elizabeth A Novak
- Department of Surgery, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Kevin P Mollen
- Department of Surgery, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| |
Collapse
|
132
|
Kautza B, Gomez H, Escobar D, Corey C, Ataya B, Luciano J, Botero AM, Gordon L, Brumfield J, Martinez S, Holder A, Ogundele O, Pinsky M, Shiva S, Zuckerbraun BS. Inhaled, nebulized sodium nitrite protects in murine and porcine experimental models of hemorrhagic shock and resuscitation by limiting mitochondrial injury. Nitric Oxide 2015; 51:7-18. [PMID: 26410351 DOI: 10.1016/j.niox.2015.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The cellular injury that occurs in the setting of hemorrhagic shock and resuscitation (HS/R) affects all tissue types and can drive altered inflammatory responses. Resuscitative adjuncts hold the promise of decreasing such injury. Here we test the hypothesis that sodium nitrite (NaNO2), delivered as a nebulized solution via an inhalational route, protects against injury and inflammation from HS/R. METHODS Mice underwent HS/R to a mean arterial pressure (MAP) of 20 or 25 mmHg. Mice were resuscitated with Lactated Ringers after 90-120 min of hypotension. Mice were randomized to receive nebulized NaNO2 via a flow through chamber (30 mg in 5 mL PBS). Pigs (30-35 kg) were anesthetized and bled to a MAP of 30-40 mmHg for 90 min, randomized to receive NaNO2 (11 mg in 2.5 mL PBS) nebulized into the ventilator circuit starting 60 min into the hypotensive period, followed by initial resuscitation with Hextend. Pigs had ongoing resuscitation and support for up to four hours. Hemodynamic data were collected continuously. RESULTS NaNO2 limited organ injury and inflammation in murine hemorrhagic shock. A nitrate/nitrite depleted diet exacerbated organ injury, as well as mortality, and inhaled NaNO2 significantly reversed this effect. Furthermore, NaNO2 limited mitochondrial oxidant injury. In porcine HS/R, NaNO2 had no significant influence on shock induced hemodynamics. NaNO2 limited hypoxia/reoxia or HS/R-induced mitochondrial injury and promoted mitochondrial fusion. CONCLUSION NaNO2 may be a useful adjunct to shock resuscitation based on its limitation of mitochondrial injury.
Collapse
Affiliation(s)
| | - Hernando Gomez
- Department of Critical Care Medicine, USA; The Center for Critical Care Nephrology, USA
| | | | | | | | | | | | | | | | | | | | | | - Michael Pinsky
- Department of Critical Care Medicine, USA; The Center for Critical Care Nephrology, USA
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, USA; Vascular Medicine Institute, University of Pittsburgh, USA.
| | - Brian S Zuckerbraun
- VA Pittsburgh Healthcare System, USA; Department of Surgery, USA; The Center for Critical Care Nephrology, USA; Vascular Medicine Institute, University of Pittsburgh, USA.
| |
Collapse
|
133
|
Gomez H, Kautza B, Escobar D, Nassour I, Luciano J, Botero AM, Gordon L, Martinez S, Holder A, Ogundele O, Loughran P, Rosengart MR, Pinsky M, Shiva S, Zuckerbraun BS. Inhaled Carbon Monoxide Protects against the Development of Shock and Mitochondrial Injury following Hemorrhage and Resuscitation. PLoS One 2015; 10:e0135032. [PMID: 26366865 PMCID: PMC4569171 DOI: 10.1371/journal.pone.0135032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 07/16/2015] [Indexed: 01/08/2023] Open
Abstract
Aims Currently, there is no effective resuscitative adjunct to fluid and blood products to limit tissue injury for traumatic hemorrhagic shock. The objective of this study was to investigate the role of inhaled carbon monoxide (CO) to limit inflammation and tissue injury, and specifically mitochondrial damage, in experimental models of hemorrhage and resuscitation. Results Inhaled CO (250 ppm for 30 minutes) protected against mortality in severe murine hemorrhagic shock and resuscitation (HS/R) (20% vs. 80%; P<0.01). Additionally, CO limited the development of shock as determined by arterial blood pH (7.25±0.06 vs. 7.05±0.05; P<0.05), lactate levels (7.2±5.1 vs 13.3±6.0; P<0.05), and base deficit (13±3.0 vs 24±3.1; P<0.05). A dose response of CO (25–500 ppm) demonstrated protection against HS/R lung and liver injury as determined by MPO activity and serum ALT, respectively. CO limited HS/R-induced increases in serum tumor necrosis factor-α and interleukin-6 levels as determined by ELISA (P<0.05 for doses of 100–500ppm). Furthermore, inhaled CO limited HS/R induced oxidative stress as determined by hepatic oxidized glutathione:reduced glutathione levels and lipid peroxidation. In porcine HS/R, CO did not influence hemodynamics. However, CO limited HS/R-induced skeletal muscle and platelet mitochondrial injury as determined by respiratory control ratio (muscle) and ATP-linked respiration and mitochondrial reserve capacity (platelets). Conclusion These preclinical studies suggest that inhaled CO can be a protective therapy in HS/R; however, further clinical studies are warranted.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Administration, Inhalation
- Animals
- Carbon Monoxide/administration & dosage
- Carbon Monoxide/pharmacology
- Carbon Monoxide/therapeutic use
- Cells, Cultured
- Interleukin-6/blood
- Lactic Acid/blood
- Male
- Mice
- Mice, Inbred C57BL
- Mitochondria, Liver/drug effects
- Mitochondria, Liver/metabolism
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/metabolism
- Oxidative Stress
- Resuscitation
- Shock, Hemorrhagic/metabolism
- Shock, Hemorrhagic/prevention & control
- Shock, Hemorrhagic/therapy
- Swine
- Tumor Necrosis Factor-alpha/blood
Collapse
Affiliation(s)
- Hernando Gomez
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- The Center for Critical Care Nephrology University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Benjamin Kautza
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Daniel Escobar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ibrahim Nassour
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jason Luciano
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ana Maria Botero
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Lisa Gordon
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Silvia Martinez
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Andre Holder
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Olufunmilayo Ogundele
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Matthew R. Rosengart
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- The Center for Critical Care Nephrology University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Michael Pinsky
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sruti Shiva
- Department of Pharmacology, University of Pittsburgh, Pittsburgh, PA, United States of America
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Brian S. Zuckerbraun
- The Center for Critical Care Nephrology University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States of America
- VA Pittsburgh Healthcare System, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
134
|
Sunasee R, Araoye E, Pyram D, Hemraz UD, Boluk Y, Ckless K. Cellulose nanocrystal cationic derivative induces NLRP3 inflammasome-dependent IL-1β secretion associated with mitochondrial ROS production. Biochem Biophys Rep 2015; 4:1-9. [PMID: 30338301 PMCID: PMC6189697 DOI: 10.1016/j.bbrep.2015.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 07/23/2015] [Accepted: 08/07/2015] [Indexed: 01/19/2023] Open
Abstract
Crystalline cellulose nanocrystals (CNCs) have emerged as novel materials for a wide variety of important applications such as nanofillers, nanocomposites, surface coatings, regenerative medicine and potential drug delivery. CNCs have a needle-like structure with sizes in the range of 100–200 nm long and 5–20 nm wide and a mean aspect ratio 10–100. Despite the great potential applicability of CNCs, very little is known about their potential immunogenicity. Needle-like materials have been known to evoke an immune response in particular to activate the (NOD-like receptor, pyrin domain-containing 3)-inflammasome/IL-1β (Interleukin 1β) pathway. In this study we evaluated the capacity of unmodified CNC and its cationic derivatives CNC-AEM (aminoethylmethacrylate)1, CNC-AEM2, CNC-AEMA(aminoethylmethacrylamide)1 and CNC-AEMA2 to stimulate NLRP3-inflammasome/IL-1β pathway and enhance the production of mitochondrial reactive oxygen species (ROS). Mouse macrophage cell line (J774A.1) was stimulated for 24 h with 50 µg/mL with unmodified CNC and its cationic derivatives. Alternatively, J774A1 or PBMCs (peripheral blood mononuclear cells) were stimulated with CNC-AEMA2 in presence or absence of LPS (lipopolysaccharide). IL-1β secretion was analyzed by ELISA, mitochondrial function by JC-1 staining and ATP content. Intracellular and mitochondrial reactive oxygen species (ROS) were assessed by DCF-DA (2′,7′-dichlorodihydrofluorescein diacetate) and MitoSOX, respectively. Mitochondrial ROS and extracellular ATP were significantly increased in cells treated with CNC-AEMA2, which correlates with the strongest effects on IL-1β secretion in non-primed cells. CNC-AEMA2 also induced IL-1βsecretion in LPS-primed and non-primed PBMCs. Our data suggest that the increases in mitochondrial ROS and ATP release induced by CNC-AEMA2 may be associated with its capability to evoke immune response. We demonstrate the first evidence that newly synthesized cationic cellulose nanocrystal derivative, CNC-AEMA2, has immunogenic properties, which may lead to the development of a potential non-toxic and safe nanomaterial to be used as a novel adjuvant for vaccines. Immunogenicity of CNC derivatives was studied on the NLRP3 inflammasome pathway. Only CNC-AEMA2 induced IL-1β secretion in non-primed and primed macrophages. IL-1β secretion caused by CNC-AEMA2 involves mitochondrial ROS and ATP release.
Collapse
Key Words
- AEM, aminoethylmethacrylate
- AEMA, aminoethylmethacrylamide
- ASC, apoptosis-associated speck-like protein containing a CARD
- CNCs, cellulose nanocrystals
- Cationic needle-like nanomaterial
- Cellulose nanocrystals
- DAMPS, danger-associated molecular pattern molecules
- DSL, Dynamic light scattering
- ELISA, enzyme-linked immuno assay
- H2DCF-DA, 2′, 7′-dichlorodihydrofluorescein diacetate
- HRP, horseradish peroxidase-conjugated
- HTCFNWs, high-temperature calcined fullerene nanowhiskers
- IL-1β
- IL-1β, Interleukin 1β
- JC-1, 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazol-carbocyanine iodide
- LPS, lipopolysaccharide
- MWCNTs, needle-like multi-wall carbon nanotubes
- Mitochondrial ROS
- Mouse macrophages
- NLRP3 inflammasome
- NLRP3, NOD-like receptor, pyrin domain-containing 3
- PAMPs, pathogen-associated molecular pattern molecules
- PBMCs, peripheral blood mononuclear cells
- ROS, reactive oxygen species
- SAA, serum amyloid A
- SDS-PAGE, sodium dodecyl sulfate polyacrylamide gel
- TMB, 3,3′,5,5′ tetramethylbezidine
Collapse
Affiliation(s)
- Rajesh Sunasee
- Department of Chemistry, State University of New York at Plattsburgh, Hudson Hall, 101 Broad Street, Plattsburgh, NY, USA
| | - Erinolaoluwa Araoye
- Department of Chemistry, State University of New York at Plattsburgh, Hudson Hall, 101 Broad Street, Plattsburgh, NY, USA
| | - Dejhy Pyram
- Department of Chemistry, State University of New York at Plattsburgh, Hudson Hall, 101 Broad Street, Plattsburgh, NY, USA
| | - Usha D Hemraz
- National Research Council of Canada, Montreal, Quebec, Canada.,Department of Civil & Environmental Engineering, University of Alberta and National Institute for Nanotechnology, National Research Council, Edmonton, Alberta, Canada
| | - Yaman Boluk
- Department of Civil & Environmental Engineering, University of Alberta and National Institute for Nanotechnology, National Research Council, Edmonton, Alberta, Canada
| | - Karina Ckless
- Department of Chemistry, State University of New York at Plattsburgh, Hudson Hall, 101 Broad Street, Plattsburgh, NY, USA
| |
Collapse
|
135
|
Zuo L, Zhou T, Pannell BK, Ziegler AC, Best TM. Biological and physiological role of reactive oxygen species--the good, the bad and the ugly. Acta Physiol (Oxf) 2015; 214:329-48. [PMID: 25912260 DOI: 10.1111/apha.12515] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/27/2015] [Accepted: 04/21/2015] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) are chemically reactive molecules that are naturally produced within biological systems. Research has focused extensively on revealing the multi-faceted and complex roles that ROS play in living tissues. In regard to the good side of ROS, this article explores the effects of ROS on signalling, immune response and other physiological responses. To review the potentially bad side of ROS, we explain the consequences of high concentrations of molecules that lead to the disruption of redox homeostasis, which induces oxidative stress damaging intracellular components. The ugly effects of ROS can be observed in devastating cardiac, pulmonary, neurodegenerative and other disorders. Furthermore, this article covers the regulatory enzymes that mitigate the effects of ROS. Glutathione peroxidase, superoxide dismutase and catalase are discussed in particular detail. The current understanding of ROS is incomplete, and it is imperative that future research be performed to understand the implications of ROS in various therapeutic interventions.
Collapse
Affiliation(s)
- L. Zuo
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
- Biophysics Graduate Program; The Ohio State University; Columbus OH USA
| | - T. Zhou
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
- Biophysics Graduate Program; The Ohio State University; Columbus OH USA
| | - B. K. Pannell
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
| | - A. C. Ziegler
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
| | - T. M. Best
- Division of Sports Medicine; Department of Family Medicine; Sports Health & Performance Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
| |
Collapse
|
136
|
Umbelliferone ameliorates cerebral ischemia-reperfusion injury via upregulating the PPAR gamma expression and suppressing TXNIP/NLRP3 inflammasome. Neurosci Lett 2015; 600:182-7. [PMID: 26071904 DOI: 10.1016/j.neulet.2015.06.016] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/05/2015] [Accepted: 06/07/2015] [Indexed: 12/23/2022]
Abstract
Umbelliferone (UMB), a natural antioxidant belonging to coumarin derivatives, is able to cross the blood-brain barrier and protect neuronal cells from death. Here we aimed to investigate the effects of UMB in a rat model of focal cerebral ischemia induced by middle cerebral artery occlusion/reperfusion (MCAO/R). Pretreatment with UMB (15 and 30 mg/kg) for 7 consecutive days ameliorated the neurological outcomes, infarct volume and brain edema in brains of MCAO rats. Our results provided evidence that UMB significantly protected neuronal cells against cerebral ischemia reperfusion-induced injury. Furthermore, UMB treatment could inhibited the level of oxidative stress and the production of inflammatory cytokines in brain tissues of MCAO rats. In addition, UMB significantly upregulated the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ), which exhibited neuroprotective effects in neurodegenerative disease. UMB treatment also suppressed NLRP3 inflammasome activation via reducing expression of Thiredoxin-interactive protein (TXNIP). These results suggest that UMB may have beneficial effects for neuroprotection against focal cerebral ischemic partly through the inhibition of TXNIP/NLRP3 inflammasome and activation of PPAR-γ.
Collapse
|
137
|
Zhang O, Ji Q, Lin Y, Wang Z, Huang Y, Lu W, Liu X, Zhang J, Liu Y, Zhou YJ. Circulating chemerin levels elevated in dilated cardiomyopathy patients with overt heart failure. Clin Chim Acta 2015; 448:27-32. [PMID: 26057200 DOI: 10.1016/j.cca.2015.05.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Recent evidence demonstrated that the circulating concentrations of adipokine are related to the presence of heart failure secondary to ischemic heart disease and dilated cardiomyopathy (DCM). However, the plasma concentrations of chemerin in patients with DCM have yet to be investigated. METHODS The present study enrolled 109 DCM patients with typical symptoms of heart failure and 60 healthy controls and measured plasma concentrations of chemerin, IL-6 and TNF-α using enzyme-linked immunosorbent assay. Left ventricular end-diastolic diameter (LVEDD) and left ventricular ejection fraction (LVEF) were measured using a GE ViVid E7 ultrasonography machine. RESULTS Plasma chemerin, IL-6 and TNF-α concentrations were significantly higher in DCM patients compared to the control group. A correlation analysis revealed that plasma chemerin concentrations were positively correlated with the concentrations of IL-6 (R=0.270, P=0.004), TNF-α (R=0.302, P=0.001), C-reactive protein (CRP) (R=0.256, P=0.004), N-terminal pro-brain natriuretic peptide (NT-proBNP) (R=0.386, P=0.000), and LVEDD (R=0.212, P=0.027) but negatively correlated with LVEF (R=-0.543, P=0.000). Furthermore, chemerin (OR 1.102, 95% CI 1.052 to 1.153; p=0.000) was independently associated with the presence of DCM before NT-proBNP was added in the multivariable regression model. CONCLUSIONS The results indicate that chemerin is a novel biomarker of DCM.
Collapse
Affiliation(s)
- Ou Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 100029, China
| | - Qingwei Ji
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 100029, China
| | - Yingzhong Lin
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Zhijian Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 100029, China
| | - Ying Huang
- Department of Ultrasound, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Wensheng Lu
- Department of Endocrinology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Xiaofei Liu
- Department of Cardiology, China-Japan Friendship Hospital of Ministry of Health, Beijing 100029, China
| | - Jianwei Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 100029, China
| | - Yuyang Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 100029, China
| | - Yu-jie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 100029, China.
| |
Collapse
|
138
|
van der Burgh R, Boes M. Mitochondria in autoinflammation: cause, mediator or bystander? Trends Endocrinol Metab 2015; 26:263-71. [PMID: 25850613 DOI: 10.1016/j.tem.2015.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/06/2015] [Accepted: 03/13/2015] [Indexed: 12/19/2022]
Abstract
People suffering from autoinflammatory disease (AID) have recurring sterile inflammation due to dysregulated inflammasome activation. Although certain genes have been associated with several AIDs, the molecular underpinnings of seemingly spontaneous inflammation are not well understood. Emerging data now suggest that mitochondrial reactive oxygen species (ROS), mitochondrial DNA (mtDNA), and autophagy might drive key signaling pathways towards activation of the inflammasome. In this review, we discuss recent findings and highlight common features between different AIDs and mitochondrial (dys)function. Although it is still early to identify clear therapeutic targets, the emerging paradigms in inflammation and mitochondrial biology show that mitochondria play an important role in AIDs, and understanding this interplay will be key in the development of new therapies.
Collapse
Affiliation(s)
- Robert van der Burgh
- Department of Pediatric Immunology and Infectious Diseases and Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht EA, 3584, The Netherlands
| | - Marianne Boes
- Department of Pediatric Immunology and Infectious Diseases and Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht EA, 3584, The Netherlands.
| |
Collapse
|
139
|
Currais A. Ageing and inflammation - A central role for mitochondria in brain health and disease. Ageing Res Rev 2015; 21:30-42. [PMID: 25684584 DOI: 10.1016/j.arr.2015.02.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 01/29/2015] [Accepted: 02/02/2015] [Indexed: 02/08/2023]
Abstract
To develop successful therapies that prevent or treat neurodegenerative diseases requires an understanding of the upstream events. Ageing is by far the greatest risk factor for most of these diseases, and to clarify their causes will require an understanding of the process of ageing itself. Starting with the question Why do we age as individual organisms, but the line of pluripotent embryonic stem cells and germ cells carried by individuals and transmitted to descendants is immortal? this review discusses how the process of cellular differentiation leads to the accumulation of biological imperfections with ageing, and how these imperfections may be the cause of chronic inflammatory responses to stress that undermine cellular function. Both differentiation and inflammation involve drastic metabolic changes associated with alterations in mitochondrial dynamics that shift the balance between aerobic glycolysis and oxidative phosphorylation. With ageing, mitochondrial dysfunction can be both the cause and consequence of inflammatory processes and elicit metabolic adaptations that might be either protective or become progressively detrimental. It is argued here that an understanding of the relationship between metabolism, differentiation and inflammation is essential to understand the pathological mechanisms governing brain health and disease during ageing.
Collapse
|
140
|
Giorgi C, Missiroli S, Patergnani S, Duszynski J, Wieckowski MR, Pinton P. Mitochondria-associated membranes: composition, molecular mechanisms, and physiopathological implications. Antioxid Redox Signal 2015; 22:995-1019. [PMID: 25557408 DOI: 10.1089/ars.2014.6223] [Citation(s) in RCA: 240] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE In all cells, the endoplasmic reticulum (ER) and mitochondria are physically connected to form junctions termed mitochondria-associated membranes (MAMs). This subcellular compartment is under intense investigation because it represents a "hot spot" for the intracellular signaling of important pathways, including the synthesis of cholesterol and phospholipids, calcium homeostasis, and reactive oxygen species (ROS) generation and activity. RECENT ADVANCES The advanced methods currently used to study this fascinating intracellular microdomain in detail have enabled the identification of the molecular composition of MAMs and their involvement within different physiopathological contexts. CRITICAL ISSUES Here, we review the knowledge regarding (i) MAMs composition in terms of protein composition, (ii) the relationship between MAMs and ROS, (iii) the involvement of MAMs in cell death programs with particular emphasis within the tumor context, (iv) the emerging role of MAMs during inflammation, and (v) the key role of MAMs alterations in selected neurological disorders. FUTURE DIRECTIONS Whether alterations in MAMs represent a response to the disease pathogenesis or directly contribute to the disease has not yet been unequivocally established. In any case, the signaling at the MAMs represents a promising pharmacological target for several important human diseases.
Collapse
Affiliation(s)
- Carlotta Giorgi
- 1 Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara , Ferrara, Italy
| | | | | | | | | | | |
Collapse
|
141
|
Pani G. Neuroprotective effects of dietary restriction: Evidence and mechanisms. Semin Cell Dev Biol 2015; 40:106-14. [DOI: 10.1016/j.semcdb.2015.03.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 12/30/2022]
|
142
|
Mechanisms by which different functional states of mitochondria define yeast longevity. Int J Mol Sci 2015; 16:5528-54. [PMID: 25768339 PMCID: PMC4394491 DOI: 10.3390/ijms16035528] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/05/2015] [Accepted: 03/05/2015] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial functionality is vital to organismal physiology. A body of evidence supports the notion that an age-related progressive decline in mitochondrial function is a hallmark of cellular and organismal aging in evolutionarily distant eukaryotes. Studies of the baker’s yeast Saccharomyces cerevisiae, a unicellular eukaryote, have led to discoveries of genes, signaling pathways and chemical compounds that modulate longevity-defining cellular processes in eukaryotic organisms across phyla. These studies have provided deep insights into mechanistic links that exist between different traits of mitochondrial functionality and cellular aging. The molecular mechanisms underlying the essential role of mitochondria as signaling organelles in yeast aging have begun to emerge. In this review, we discuss recent progress in understanding mechanisms by which different functional states of mitochondria define yeast longevity, outline the most important unanswered questions and suggest directions for future research.
Collapse
|
143
|
Itoh K, Ye P, Matsumiya T, Tanji K, Ozaki T. Emerging functional cross-talk between the Keap1-Nrf2 system and mitochondria. J Clin Biochem Nutr 2015; 56:91-7. [PMID: 25759513 PMCID: PMC4345178 DOI: 10.3164/jcbn.14-134] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/01/2014] [Indexed: 12/30/2022] Open
Abstract
Nuclear factor erythroid-derived 2-related factor 2 (Nrf2) was originally identified as a positive regulator of drug detoxifying enzyme gene expression during exposure to environmental electrophiles. Currently, Nrf2 is known to regulate the expression of hundreds of cytoprotective genes to counteract endogenously or exogenously generated oxidative stress. Furthermore, when activated in human tumors by somatic mutations, Nrf2 confers growth advantages and chemoresistance by regulating genes involved in various processes such as the pentose phosphate pathway and nucleotide synthesis in addition to antioxidant proteins. Interestingly, increasing evidence shows that Nrf2 is associated with mitochondrial biogenesis during environmental stresses in certain tissues such as the heart. Furthermore, SKN-1, a functional homolog of Nrf2 in C. elegans, is activated by mitochondrial reactive oxygen species and extends life span by promoting mitochondrial homeostasis (i.e., mitohormesis). Similarly, Nrf2 activation was recently observed in the heart of surfeit locus protein 1 (Surf1) -/- mice in which cellular respiration was decreased due to cytochrome c oxidase defects. In this review, we critically examine the relationship between Nrf2 and mitochondria and argue that the Nrf2 stress pathway intimately communicates with mitochondria to maintain cellular homeostasis during oxidative stress.
Collapse
Affiliation(s)
- Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan
| | - Peng Ye
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan
| | - Tomoh Matsumiya
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan
| | - Taku Ozaki
- Research Center for Child Mental Development, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan
| |
Collapse
|
144
|
Coelho C, Souza ACO, Derengowski LDS, de Leon-Rodriguez C, Wang B, Leon-Rivera R, Bocca AL, Gonçalves T, Casadevall A. Macrophage mitochondrial and stress response to ingestion of Cryptococcus neoformans. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:2345-57. [PMID: 25646306 PMCID: PMC4340727 DOI: 10.4049/jimmunol.1402350] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human infection with Cryptococcus neoformans, a common fungal pathogen, follows deposition of yeast spores in the lung alveoli. The subsequent host-pathogen interaction can result in eradication, latency, or extrapulmonary dissemination. Successful control of C. neoformans infection is dependent on host macrophages, but macrophages display little ability to kill C. neoformans in vitro. Recently, we reported that ingestion of C. neoformans by mouse macrophages induces early cell cycle progression followed by mitotic arrest, an event that almost certainly reflects host cell damage. The goal of the present work was to understand macrophage pathways affected by C. neoformans toxicity. Infection of macrophages by C. neoformans was associated with alterations in protein translation rate and activation of several stress pathways, such as hypoxia-inducing factor-1-α, receptor-interacting protein 1, and apoptosis-inducing factor. Concomitantly we observed mitochondrial depolarization in infected macrophages, an observation that was replicated in vivo. We also observed differences in the stress pathways activated, depending on macrophage cell type, consistent with the nonspecific nature of C. neoformans virulence known to infect phylogenetically distant hosts. Our results indicate that C. neoformans infection impairs multiple host cellular functions and undermines the health of these critical phagocytic cells, which can potentially interfere with their ability to clear this fungal pathogen.
Collapse
Affiliation(s)
- Carolina Coelho
- Department of Microbiology and Immunology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461; Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Camila Oliveira Souza
- Cell Biology Department, Biology Science Institute, University of Brasilia, Brasilia CEP 70910-900, Brazil
| | | | - Carlos de Leon-Rodriguez
- Department of Microbiology and Immunology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Bo Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461; MD Program, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Rosiris Leon-Rivera
- Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico 00931; and Undergraduate Research Program, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Anamelia Lorenzetti Bocca
- Cell Biology Department, Biology Science Institute, University of Brasilia, Brasilia CEP 70910-900, Brazil
| | - Teresa Gonçalves
- Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Arturo Casadevall
- Department of Microbiology and Immunology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461;
| |
Collapse
|
145
|
Abstract
In addition to their role as energy generators, mitochondria play critical and active roles in diverse signalling pathways, from immunity to cell survival and cell fate decisions. However, there remain many open questions and challenges as we work towards integrating this mighty organelle into established paradigms of cellular physiology.
Collapse
Affiliation(s)
- Heidi M McBride
- Montreal Neurological Institute, McGill University, 3801 University Avenue, Rm 622C H3A 2B4, Montreal H3A 0G4, QC, Canada.
| |
Collapse
|
146
|
Mitochondrial Ca2+-dependent NLRP3 activation exacerbates the Pseudomonas aeruginosa-driven inflammatory response in cystic fibrosis. Nat Commun 2015; 6:6201. [PMID: 25648527 DOI: 10.1038/ncomms7201] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/05/2015] [Indexed: 12/28/2022] Open
Abstract
The common pathological manifestation of cystic fibrosis (CF) is associated with an excessive lung inflammatory response characterized by interleukin-1β accumulation. CF airway epithelial cells show an exacerbated pro-inflammatory response to Pseudomonas aeruginosa; however, it is unclear whether this heightened inflammatory response is intrinsic to cells lacking CF transmembrane conductance regulator (CFTR). Here we demonstrate that the degree and quality of the inflammatory response in CF are supported by P. aeruginosa-dependent mitochondrial perturbation, in which flagellin is the inducer and mitochondrial Ca(2+) uniporter (MCU) is a signal-integrating organelle member for NLRP3 activation and IL-1β and IL-18 processing. Our work elucidates the regulation of the NLRP3 inflammasome by mitochondrial Ca(2+) in the P. aeruginosa-dependent inflammatory response and deepens our understanding of the significance of mitochondria in the Ca(2+)-dependent control of inflammation.
Collapse
|
147
|
Deciphering of mitochondrial cardiolipin oxidative signaling in cerebral ischemia-reperfusion. J Cereb Blood Flow Metab 2015; 35:319-28. [PMID: 25407268 PMCID: PMC4426750 DOI: 10.1038/jcbfm.2014.204] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 11/08/2022]
Abstract
It is believed that biosynthesis of lipid mediators in the central nervous system after cerebral ischemia-reperfusion starts with phospholipid hydrolysis by calcium-dependent phospholipases and is followed by oxygenation of released fatty acids (FAs). Here, we report an alternative pathway whereby cereberal ischemia-reperfusion triggered oxygenation of a mitochondria-specific phospholipid, cardiolipin (CL), is followed by its hydrolysis to yield monolyso-CLs and oxygenated derivatives of fatty (linoleic) acids. We used a model of global cerebral ischemia-reperfusion characterized by 9 minutes of asphyxia leading to asystole followed by cardiopulmonary resuscitation in postnatal day 17 rats. Global ischemia and cardiopulmonary resuscitation resulted in: (1) selective oxidation and hydrolysis of CLs, (2) accumulation of lyso-CLs and oxygenated free FAs, (3) activation of caspase 3/7 in the brain, and (4) motor and cognitive dysfunction. On the basis of these findings, we used a mitochondria targeted nitroxide electron scavenger, which prevented CL oxidation and subsequent hydrolysis, attenuated caspase activation, and improved neurocognitive outcome when administered after cardiac arrest. These data show that calcium-independent CL oxidation and subsequent hydrolysis represent a previously unidentified pathogenic mechanism of brain injury incurred by ischemia-reperfusion and a clinically relevant therapeutic target.
Collapse
|
148
|
Dash P, Thomas PG. Host detection and the stealthy phenotype in influenza virus infection. Curr Top Microbiol Immunol 2015; 386:121-47. [PMID: 25038940 DOI: 10.1007/82_2014_412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The innate host response to influenza virus infection plays a critical role in determining the subsequent course of infection and the clinical outcome of disease. The host has a diverse array of detection and effector mechanisms that are able to recognize and initiate effective antiviral responses. In opposition, the virus utilizes a number of distinct mechanisms to evade host detection and effector activity in order to remain "stealthy" throughout its replication cycle. In this review, we describe these host and viral mechanisms, including the major pattern recognition receptor families (the TLRs, NLRs, and RLRs) in the host and the specific viral proteins such as NS1 that are key players in this interaction. Additionally, we explore nonreductive mechanisms of viral immune evasion and propose areas important for future inquiry.
Collapse
Affiliation(s)
- Pradyot Dash
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | |
Collapse
|
149
|
Abstract
Chronic, low-grade inflammation in osteoarthritis (OA) contributes to symptoms and disease progression. Effective disease-modifying OA therapies are lacking, but better understanding inflammatory pathophysiology in OA could lead to transformative therapy. Networks of diverse innate inflammatory danger signals, including complement and alarmins, are activated in OA. Through inflammatory mediators, biomechanical injury and oxidative stress compromise the viability of chondrocytes, reprogramming them to hypertrophic differentiation and proinflammatory and pro-catabolic responses. Integral to this reprogramming are 'switching' pathways in transcriptional networks, other than the well-characterized effects of NFκB and mitogen-activated protein kinase signalling; HIF-2α transcriptional signalling and ZIP8-mediated Zn(2+) uptake, with downstream MTF1 transcriptional signalling, have been implicated but further validation is required. Permissive factors, including impaired bioenergetics via altered mitochondrial function and decreased activity of bioenergy sensors, interact with molecular inflammatory responses and proteostasis mechanisms such as the unfolded protein response and autophagy. Bioenergy-sensing by AMPK and SIRT1 provides 'stop signals' for oxidative stress, inflammatory, and matrix catabolic processes in chondrocytes. The complexity of molecular inflammatory processes in OA and the involvement of multiple inflammatory mediators in tissue repair responses, raises daunting questions about how to therapeutically target inflammatory processes and macroscopic inflammation in OA. Bioenergy sensing might provide a pragmatic 'entry point'.
Collapse
Affiliation(s)
- Ru Liu-Bryan
- San Diego VA Healthcare System and Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 111K, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| | - Robert Terkeltaub
- San Diego VA Healthcare System and Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 111K, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| |
Collapse
|
150
|
Ortiz F, Acuña-Castroviejo D, Doerrier C, Dayoub JC, López LC, Venegas C, García JA, López A, Volt H, Luna-Sánchez M, Escames G. Melatonin blunts the mitochondrial/NLRP3 connection and protects against radiation-induced oral mucositis. J Pineal Res 2015; 58:34-49. [PMID: 25388914 DOI: 10.1111/jpi.12191] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/07/2014] [Indexed: 12/13/2022]
Abstract
Mucositis is a common and distressing side effect of chemotherapy or radiotherapy that has potentially severe consequences, and no treatment is available. The purpose of this study was to analyze the molecular pathways involved in the development of oral mucositis and to evaluate whether melatonin can prevent this pathology. The tongue of male Wistar rats was subjected to irradiation (X-ray YXLON Y.Tu 320-D03 irradiator; the animals received a dose of 7.5 Gy/day for 5 days). Rats were treated with 45 mg/day melatonin or vehicle for 21 days postirradiation, either by local application into their mouths (melatonin gel) or by subcutaneous injection. A connection between reactive oxygen species, generating mitochondria and the NLRP3 (NLR-related protein 3 nucleotide-binding domain leucine-rich repeat containing receptor-related protein 3) inflammasome, has been reported in mucositis. Here, we show that mitochondrial oxidative stress, bioenergetic impairment and subsequent NLRP3 inflammasome activation are involved in the development of oral mucositis after irradiation and that melatonin synthesized in the rat tongue is depleted after irradiation. The application of melatonin gel restores physiological melatonin levels in the tongue and prevents mucosal disruption and ulcer formation. Melatonin gel protects the mitochondria from radiation damage and blunts the NF-κB/NLRP3 inflammasome signaling activation in the tongue. Our results suggest new molecular pathways involved in radiotherapy-induced mucositis that are inhibited by topical melatonin application, suggesting a potential preventive therapy for mucositis in patients with cancer.
Collapse
Affiliation(s)
- Francisco Ortiz
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, and RETICEF, Granada, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|