1
|
De Nicola AF, Meyer M, Garay L, Kruse MS, Schumacher M, Guennoun R, Gonzalez Deniselle MC. Progesterone and Allopregnanolone Neuroprotective Effects in the Wobbler Mouse Model of Amyotrophic Lateral Sclerosis. Cell Mol Neurobiol 2021; 42:23-40. [PMID: 34138412 DOI: 10.1007/s10571-021-01118-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023]
Abstract
Progesterone regulates a number of processes in neurons and glial cells not directly involved in reproduction or sex behavior. Several neuroprotective effects are better observed under pathological conditions, as shown in the Wobbler mouse model of amyotrophic laterals sclerosis (ALS). Wobbler mice are characterized by forelimb atrophy due to motoneuron degeneration in the spinal cord, and include microgliosis and astrogliosis. Here we summarized current evidence on progesterone reversal of Wobbler neuropathology. We demonstrated that progesterone decreased motoneuron vacuolization with preservation of mitochondrial respiratory complex I activity, decreased mitochondrial expression and activity of nitric oxide synthase, increased Mn-dependent superoxide dismutase, stimulated brain-derived neurotrophic factor, increased the cholinergic phenotype of motoneurons, and enhanced survival with a concomitant decrease of death-related pathways. Progesterone also showed differential effects on glial cells, including increased oligodendrocyte density and downregulation of astrogliosis and microgliosis. These changes associate with reduced anti-inflammatory markers. The enhanced neurochemical parameters were accompanied by longer survival and increased muscle strength in tests of motor behavior. Because progesterone is locally metabolized to allopregnanolone (ALLO) in nervous tissues, we also studied neuroprotection by this derivative. Treatment of Wobbler mice with ALLO decreased oxidative stress and glial pathology, increased motoneuron viability and clinical outcome in a progesterone-like manner, suggesting that ALLO could mediate some progesterone effects in the spinal cord. In conclusion, the beneficial effects observed in different parameters support the versatile properties of progesterone and ALLO in a mouse model of motoneuron degeneration. The studies foresee future therapeutic opportunities with neuroactive steroids for deadly diseases like ALS.
Collapse
Affiliation(s)
- Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina. .,Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425, Buenos Aires, Argentina.
| | - María Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Laura Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina.,Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425, Buenos Aires, Argentina
| | - Maria Sol Kruse
- Laboratory of Neurobiology, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Michael Schumacher
- U1195 INSERM and University Paris Sud "Neuroprotective, Neuroregenerative and Remyelinating Small Molecules, 94276, Kremlin-Bicetre, France
| | - Rachida Guennoun
- U1195 INSERM and University Paris Sud "Neuroprotective, Neuroregenerative and Remyelinating Small Molecules, 94276, Kremlin-Bicetre, France
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina.,Department of Physiological Sciences, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425, Buenos Aires, Argentina
| |
Collapse
|
2
|
Gheibi S, Ghasemi A. Insulin secretion: The nitric oxide controversy. EXCLI JOURNAL 2020; 19:1227-1245. [PMID: 33088259 PMCID: PMC7573190 DOI: 10.17179/excli2020-2711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022]
Abstract
Nitric oxide (NO) is a gas that serves as a ubiquitous signaling molecule participating in physiological activities of various organ systems. Nitric oxide is produced in the endocrine pancreas and contributes to synthesis and secretion of insulin. The potential role of NO in insulin secretion is disputable - both stimulatory and inhibitory effects have been reported. Available data indicate that effects of NO critically depend on its concentration. Different isoforms of NO synthase (NOS) control this and have the potential to decrease or increase insulin secretion. In this review, the role of NO in insulin secretion as well as the possible reasons for discrepant findings are discussed. A better understanding of the role of NO system in the regulation of insulin secretion may facilitate the development of new therapeutic strategies in the management of diabetes.
Collapse
Affiliation(s)
- Sevda Gheibi
- Department of Clinical Sciences in Malmö, Unit of Molecular Metabolism, Lund University Diabetes Centre, Clinical Research Center, Malmö University Hospital, Lund University, Malmö, Sweden
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Stožer A, Vodopivc P, Križančić Bombek L. Pathophysiology of exercise-induced muscle damage and its structural, functional, metabolic, and clinical consequences. Physiol Res 2020; 69:565-598. [PMID: 32672048 DOI: 10.33549/physiolres.934371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Extreme or unaccustomed eccentric exercise can cause exercise-induced muscle damage, characterized by structural changes involving sarcomere, cytoskeletal, and membrane damage, with an increased permeability of sarcolemma for proteins. From a functional point of view, disrupted force transmission, altered calcium homeostasis, disruption of excitation-contraction coupling, as well as metabolic changes bring about loss of strength. Importantly, the trauma also invokes an inflammatory response and clinically presents itself by swelling, decreased range of motion, increased passive tension, soreness, and a transient decrease in insulin sensitivity. While being damaging and influencing heavily the ability to perform repeated bouts of exercise, changes produced by exercise-induced muscle damage seem to play a crucial role in myofibrillar adaptation. Additionally, eccentric exercise yields greater hypertrophy than isometric or concentric contractions and requires less in terms of metabolic energy and cardiovascular stress, making it especially suitable for the elderly and people with chronic diseases. This review focuses on our current knowledge of the mechanisms underlying exercise-induced muscle damage, their dependence on genetic background, as well as their consequences at the structural, functional, metabolic, and clinical level. A comprehensive understanding of these is a prerequisite for proper inclusion of eccentric training in health promotion, rehabilitation, and performance enhancement.
Collapse
Affiliation(s)
- A Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Slovenia.
| | | | | |
Collapse
|
4
|
Ramachandran A, Jaeschke H. A mitochondrial journey through acetaminophen hepatotoxicity. Food Chem Toxicol 2020; 140:111282. [PMID: 32209353 PMCID: PMC7254872 DOI: 10.1016/j.fct.2020.111282] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022]
Abstract
Acetaminophen (APAP) overdose is the leading cause of acute liver failure in the United States and APAP-induced hepatotoxicity is initiated by formation of a reactive metabolite which depletes hepatic glutathione and forms protein adducts. Studies over the years have established the critical role of c-Jun N terminal kinase (JNK) and its mitochondrial translocation, as well as mitochondrial oxidant stress and subsequent induction of the mitochondrial permeability transition in APAP pathophysiology. However, it is now evident that mitochondrial responses to APAP overdose are more nuanced than appreciated earlier, with multiple levels of control, for example, to dose of APAP. In addition, mitochondrial dynamics, as well as the organelle's importance in recovery and regeneration after APAP-induced liver injury is also being recognized, which are exciting new areas with significant therapeutic potential. Thus, this review examines the temporal course of hepatocyte mitochondrial responses to an APAP overdose with an emphasis on mechanistic response to various trigger checkpoints such as NAPQI-mitochondrial protein adduct formation and activated JNK translocation. Mitochondrial dynamics, the organelle's role in recovery after APAP and emerging areas of research which promise to provide further insight into modulation of APAP pathophysiology by these fascinating organelles will also be discussed.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology, and Therapeutic, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology, and Therapeutic, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
5
|
Gheibi S, Samsonov AP, Gheibi S, Vazquez AB, Kashfi K. Regulation of carbohydrate metabolism by nitric oxide and hydrogen sulfide: Implications in diabetes. Biochem Pharmacol 2020; 176:113819. [PMID: 31972170 DOI: 10.1016/j.bcp.2020.113819] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/15/2020] [Indexed: 12/19/2022]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are two gasotransmitters that are produced in the human body and have a key role in many of the physiological activities of the various organ systems. Decreased NO bioavailability and deficiency of H2S are involved in the pathophysiology of type 2 diabetes and its complications. Restoration of NO levels have favorable metabolic effects in diabetes. The role of H2S in pathophysiology of diabetes is however controversial; H2S production is decreased during development of obesity, diabetes, and its complications, suggesting the potential therapeutic effects of H2S. On the other hand, increased H2S levels disturb the pancreatic β-cell function and decrease insulin secretion. In addition, there appear to be important interactions between NO and H2S at the levels of both biosynthesis and signaling pathways, yet clear an insight into this relationship is lacking. H2S potentiates the effects of NO in the cardiovascular system as well as NO release from its storage pools. Likewise, NO increases the activity and the expression of H2S-generating enzymes. Inhibition of NO production leads to elimination/attenuation of the cardioprotective effects of H2S. Regarding the increasing interest in the therapeutic applications of NO or H2S-releasing molecules in a variety of diseases, particularly in the cardiovascular disorders, much is to be learned about their function in glucose/insulin metabolism, especially in diabetes. The aim of this review is to provide a better understanding of the individual and the interactive roles of NO and H2S in carbohydrate metabolism.
Collapse
Affiliation(s)
- Sevda Gheibi
- Department of Clinical Sciences in Malmö, Unit of Molecular Metabolism, Lund University Diabetes Centre, Clinical Research Center, Malmö University Hospital, Lund University, Malmö, Sweden.
| | - Alan P Samsonov
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Shahsanam Gheibi
- Maternal and Childhood Obesity Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Alexandra B Vazquez
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, NY, USA.
| |
Collapse
|
6
|
The Comparative Cytotoxic Effects of Apis mellifera Crude Venom on MCF-7 Breast Cancer Cell Line in 2D and 3D Cell Cultures. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09979-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
7
|
De Armas MI, Esteves R, Viera N, Reyes AM, Mastrogiovanni M, Alegria TGP, Netto LES, Tórtora V, Radi R, Trujillo M. Rapid peroxynitrite reduction by human peroxiredoxin 3: Implications for the fate of oxidants in mitochondria. Free Radic Biol Med 2019; 130:369-378. [PMID: 30391677 DOI: 10.1016/j.freeradbiomed.2018.10.451] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Mitochondria are main sites of peroxynitrite formation. While at low concentrations mitochondrial peroxynitrite has been associated with redox signaling actions, increased levels can disrupt mitochondrial homeostasis and lead to pathology. Peroxiredoxin 3 is exclusively located in mitochondria, where it has been previously shown to play a major role in hydrogen peroxide reduction. In turn, reduction of peroxynitrite by peroxiredoxin 3 has been inferred from its protective actions against tyrosine nitration and neurotoxicity in animal models, but was not experimentally addressed so far. Herein, we demonstrate the human peroxiredoxin 3 reduces peroxynitrite with a rate constant of 1 × 107 M-1 s-1 at pH 7.8 and 25 °C. Reaction with hydroperoxides caused biphasic changes in the intrinsic fluorescence of peroxiredoxin 3: the first phase corresponded to the peroxidatic cysteine oxidation to sulfenic acid. Peroxynitrite in excess led to peroxiredoxin 3 hyperoxidation and tyrosine nitration, oxidative post-translational modifications that had been previously identified in vivo. A significant fraction of the oxidant is expected to react with CO2 and generate secondary radicals, which participate in further oxidation and nitration reactions, particularly under metabolic conditions of active oxidative decarboxylations or increased hydroperoxide formation. Our results indicate that both peroxiredoxin 3 and 5 should be regarded as main targets for peroxynitrite in mitochondria.
Collapse
Affiliation(s)
- María Inés De Armas
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay
| | - Romina Esteves
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay
| | - Nicolás Viera
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay
| | - Aníbal M Reyes
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay
| | - Mauricio Mastrogiovanni
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay
| | - Thiago G P Alegria
- Departamento de Genética e Biología Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biología Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Verónica Tórtora
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Uruguay; Center For Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Uruguay.
| |
Collapse
|
8
|
Nitric Oxide and Mitochondrial Function in Neurological Diseases. Neuroscience 2018; 376:48-71. [DOI: 10.1016/j.neuroscience.2018.02.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/20/2018] [Accepted: 02/09/2018] [Indexed: 12/17/2022]
|
9
|
Zhang H, Wang Q, Gu J, Yin L, Liang S, Wu L, Xu H, Zhao C, Gu Y. Elevated mitochondrial SLC25A29 in cancer modulates metabolic status by increasing mitochondria-derived nitric oxide. Oncogene 2018; 37:2545-2558. [PMID: 29459713 DOI: 10.1038/s41388-018-0139-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/17/2017] [Accepted: 12/22/2017] [Indexed: 11/09/2022]
Abstract
Warburg effect has been recognized as a hallmark of cancer cells for many years, but its modulation mechanism remains a great focus. Our current study found a member of solute carrier family 25 (SLC25A29), the main arginine transporter on mitochondria, significantly elevated in various cancer cells. Knockout of SLC25A29 by CRISPR/Cas9 inhibited proliferation and migration of cancer cells both in vitro and in vivo. SLC25A29-knockout cells also showed an altered metabolic status with enhanced mitochondrial respiration and reduced glycolysis. All of above impacts could be reversed after rescuing SLC25A29 expression in SLC25A29-knockout cells. Arginine is transported into mitochondria partly for nitric oxide (NO) synthesis. Deletion of SLC25A29 resulted in severe decrease of NO production, indicating that the mitochondria is a significant source of NO. SLC25A29-knockout cells dramatically altered the variation of metabolic processes, whereas addition of arginine failed to reverse the effect, highlighting the necessity of transporting arginine into mitochondria by SLC25A29. In conclusion, aberrant elevated SLC25A29 in cancer functioned to transport more arginine into mitochondria, improved mitochondria-derived NO levels, thus modulated metabolic status to facilitate increased cancer progression.
Collapse
Affiliation(s)
- Huiyuan Zhang
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Qinyi Wang
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Junzhong Gu
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Le Yin
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Shenghui Liang
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Lida Wu
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Hao Xu
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Chao Zhao
- Department of Clinical Neurosciences, WT-MRC Cambridge Stem Cell Institute, Cambridge University, Cambridge, UK
| | - Yuchun Gu
- Laboratory of Molecular Pharmacology, Institute of Molecular Medicine, Peking University, Beijing, China. .,Translational and Regenerative Medicine Center, Aston Medical Research Institute, Aston University, Birmingham, UK.
| |
Collapse
|
10
|
Beaulieu L, Vitseva O, Tanriverdi K, Kucukural A, Mick E, Hamburg N, Vita J, Freedman J. Platelet functional and transcriptional changes induced by intralipid infusion. Thromb Haemost 2017; 115:1147-56. [DOI: 10.1160/th15-09-0739] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/11/2016] [Indexed: 02/07/2023]
Abstract
SummaryMultiple studies have shown the effects of long-term exposure to high-fat or western diets on the vascular system. There is limited knowledge on the acute effects of high circulating fat levels, specifically on platelets, which have a role in many processes, including thrombosis and inflammation. This study investigated the effects of acute, high-fat exposure on platelet function and transcript profile. Twenty healthy participants were given an intravenous infusion of 20% Intralipid emulsion and heparin over 6 hours. Blood samples were taken prior to and the day after infusion to measure platelet function and transcript expression levels. Platelet aggregation was not significantly affected by Intralipid infusion, but, when mitochondria function was inhibited by carbonyl cyanide 3-chlorophenylhydrazone (CCCP) or oligomycin, platelet aggregation was higher in the post-infusion state compared to baseline. Through RNA sequencing, and verified by RT-qPCR, 902 miRNAs and 617 mRNAs were affected by Intralipid infusion. MicroRNAs increased include miR-4259 and miR-346, while miR-517b and miR-517c are both decreased. Pathway analysis identified two clusters significantly enriched, including cell motility. In conclusion, acute exposure to high fat affects mitochondrial-dependent platelet function, as well as the transcript profile.
Collapse
|
11
|
Susilo I, Devi A, Purwandhono A, Warsito SH. Effects of hyperbaric oxygen therapy in enhancing expressions of e-NOS, TNF-α and VEGF in wound healing. ACTA ACUST UNITED AC 2017. [DOI: 10.1088/1742-6596/853/1/012030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
12
|
Resveratrol and Brain Mitochondria: a Review. Mol Neurobiol 2017; 55:2085-2101. [DOI: 10.1007/s12035-017-0448-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/07/2017] [Indexed: 12/24/2022]
|
13
|
Pienaar IS, Vernon A, Winn P. The Cellular Diversity of the Pedunculopontine Nucleus: Relevance to Behavior in Health and Aspects of Parkinson's Disease. Neuroscientist 2016; 23:415-431. [PMID: 27932591 DOI: 10.1177/1073858416682471] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The pedunculopontine nucleus (PPN) is a rostral brainstem structure that has extensive connections with basal ganglia nuclei and the thalamus. Through these the PPN contributes to neural circuits that effect cortical and hippocampal activity. The PPN also has descending connections to nuclei of the pontine and medullary reticular formations, deep cerebellar nuclei, and the spinal cord. Interest in the PPN has increased dramatically since it was first suggested to be a novel target for treating patients with Parkinson's disease who are refractory to medication. However, application of frequency-specific electrical stimulation of the PPN has produced inconsistent results. A central reason for this is that the PPN is not a heterogeneous structure. In this article, we review current knowledge of the neurochemical identity and topographical distribution of neurons within the PPN of both humans and experimental animals, focusing on studies that used neuronally selective targeting strategies to ascertain how the neurochemical heterogeneity of the PPN relates to its diverse functions in relation to movement and cognitive processes. If the therapeutic potential of the PPN is to be realized, it is critical to understand the complex structure-function relationships that exist here.
Collapse
Affiliation(s)
- Ilse S Pienaar
- 1 Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Cane Road, London, UK.,2 Faculty of Health and Life Sciences, Department of Applied Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Anthony Vernon
- 3 Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Philip Winn
- 4 Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
14
|
Kovaleva VD, Uzdensky AB. Photodynamic therapy-induced nitric oxide production in neuronal and glial cells. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:105005. [PMID: 27784050 DOI: 10.1117/1.jbo.21.10.105005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/05/2016] [Indexed: 06/06/2023]
Abstract
Nitric oxide (NO) has been recently demonstrated to enhance apoptosis of glial cells induced by photodynamic therapy (PDT), but to protect glial cells from PDT-induced necrosis in the crayfish stretch receptor, a simple neuroglial preparation that consists of a single mechanosensory neuron enveloped by satellite glial cells. We used the NO-sensitive fluorescent probe 4,5-diaminofluorescein diacetate to study the distribution and dynamics of PDT-induced NO production in the mechanosensory neuron and surrounding glial cells. The NO production in the glial envelope was higher than in the neuronal soma axon and dendrites both in control and in experimental conditions. In dark NO generator, DEA NONOate or NO synthase substrate L-arginine hydrochloride significantly increased the NO level in glial cells, whereas NO scavenger 2-Phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (PTIO) or inhibitors of NO synthase L-NG-nitro arginine methyl ester and N?-nitro-L-arginine decreased it. PDT induced the transient increase in NO production with a maximum at 4 to 7 min after the irradiation start followed by its inhibition at 10 to 40 min. We suggested that PDT stimulated neuronal rather than inducible NO synthase isoform in glial cells, and the produced NO could mediate PDT-induced apoptosis.
Collapse
Affiliation(s)
- Vera D Kovaleva
- Southern Federal University, Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Stachky Avenue 194/1, 344090 Rostov-on-Don, Russia
| | - Anatoly B Uzdensky
- Southern Federal University, Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Stachky Avenue 194/1, 344090 Rostov-on-Don, Russia
| |
Collapse
|
15
|
Sarfo-Poku C, Eshun O, Lee KH. Medical application of scorpion venom to breast cancer: A mini-review. Toxicon 2016; 122:109-112. [PMID: 27644898 DOI: 10.1016/j.toxicon.2016.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 04/21/2016] [Accepted: 09/06/2016] [Indexed: 12/22/2022]
Abstract
Breast cancer is the leading cause of mortality in women worldwide. Today, 1 in 8 women born in the United States will have an invasive cancer in their lifetime. Despite significant attempts, the prognosis of metastatic breast cancer still remains poor. This has compelled scientists to look elsewhere for better therapeutic outcomes. Recent advances in venomic studies have demonstrated some promise in cancer-related ailments. Scorpion venom, a complex cocktail of biogenic amines, proteins, peptides, mucoproteins, organic salts and neurotoxins has shown a potential therapeutic application due to its cytotoxic, apoptogenic, immunosuppressive and antiproliferative properties. This communication reviews the effects of scorpion venom components on breast cancer and their mechanisms.
Collapse
Affiliation(s)
- Christian Sarfo-Poku
- Department of Earth & Biological Sciences, School of Medicine Loma Linda University, Loma Linda, CA 92350, United States.
| | - Oliver Eshun
- Department of Earth & Biological Sciences, School of Medicine Loma Linda University, Loma Linda, CA 92350, United States.
| | - Kyung Hyun Lee
- Department of Earth & Biological Sciences, School of Medicine Loma Linda University, Loma Linda, CA 92350, United States
| |
Collapse
|
16
|
Zubeldia-Brenner L, Roselli CE, Recabarren SE, Gonzalez Deniselle MC, Lara HE. Developmental and Functional Effects of Steroid Hormones on the Neuroendocrine Axis and Spinal Cord. J Neuroendocrinol 2016; 28:10.1111/jne.12401. [PMID: 27262161 PMCID: PMC4956521 DOI: 10.1111/jne.12401] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 06/03/2016] [Accepted: 06/03/2016] [Indexed: 12/13/2022]
Abstract
This review highlights the principal effects of steroid hormones at central and peripheral levels in the neuroendocrine axis. The data discussed highlight the principal role of oestrogens and testosterone in hormonal programming in relation to sexual orientation, reproductive and metabolic programming, and the neuroendocrine mechanism involved in the development of polycystic ovary syndrome phenotype. Moreover, consistent with the wide range of processes in which steroid hormones take part, we discuss the protective effects of progesterone on neurodegenerative disease and the signalling mechanism involved in the genesis of oestrogen-induced pituitary prolactinomas.
Collapse
Affiliation(s)
- L Zubeldia-Brenner
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - C E Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University Portland, Portland, OR, USA
| | - S E Recabarren
- Laboratory of Animal Physiology and Endocrinology, Faculty of Veterinary Sciences, University of Concepcion, Chillán, Chile
| | - M C Gonzalez Deniselle
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - H E Lara
- Laboratory of Neurobiochemistry Department of Biochemistry and Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|
17
|
Kitamura K, Kawaguchi M, Ieda N, Miyata N, Nakagawa H. Visible Light-Controlled Nitric Oxide Release from Hindered Nitrobenzene Derivatives for Specific Modulation of Mitochondrial Dynamics. ACS Chem Biol 2016; 11:1271-8. [PMID: 26878937 DOI: 10.1021/acschembio.5b00962] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is a physiological signaling molecule, whose biological production is precisely regulated at the subcellular level. Here, we describe the design, synthesis, and evaluation of novel mitochondria-targeted NO releasers, Rol-DNB-mor and Rol-DNB-pyr, that are photocontrollable not only in the UV wavelength range but also in the biologically favorable visible wavelength range (530-590 nm). These caged NO compounds consist of a hindered nitrobenzene as the NO-releasing moiety and a rhodamine chromophore. Their NO-release properties were characterized by an electron spin resonance (ESR) spin trapping method and fluorometric analysis using NO probes, and their mitochondrial localization in live cells was confirmed by costaining. Furthermore, we demonstrated visible light control of mitochondrial fragmentation via activation of dynamin-related protein 1 (Drp1) by means of precisely controlled NO delivery into mitochondria of cultured HEK293 cells, utilizing Rol-DNB-pyr.
Collapse
Affiliation(s)
- Kai Kitamura
- Graduate School of Pharmaceutical
Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Mitsuyasu Kawaguchi
- Graduate School of Pharmaceutical
Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Naoya Ieda
- Graduate School of Pharmaceutical
Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Naoki Miyata
- Graduate School of Pharmaceutical
Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Hidehiko Nakagawa
- Graduate School of Pharmaceutical
Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| |
Collapse
|
18
|
Chemopreventive effect of 18β-glycyrrhetinic acid via modulation of inflammatory markers and induction of apoptosis in human hepatoma cell line (HepG2). Mol Cell Biochem 2016; 416:169-77. [PMID: 27116616 DOI: 10.1007/s11010-016-2705-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/13/2016] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma is one of the most common lethal diseases worldwide and there is no effective treatment till date. Natural products derived from the plants play an important role in chemoprevention and act as therapeutic antitumor agents. Licorice is a plant that has been used in food and medicine for the treatment of various diseases. 18β-Glycyrrhetinic acid (18β-GA), a pentacyclic triterpenoid obtained from the roots of licorice plant, is reported to possess various pharmacological properties such as antitumor and antiinflammatory activities. The present study was designed to elucidate the chemopreventive effect of 18β-GA through antiinflammation, antiproliferation, and induction of apoptosis in human hepatoma cell line HepG2. 18β-GA significantly inhibits the proliferation of HepG2 cell without affecting the normal liver cell line (Chang's). In the present study, 18β-GA increased the formation of reactive oxygen species, nitric oxide production, and loss of mitochondrial membrane potential, suggesting the involvement of 18β-GA in apoptosis which was also confirmed by assessing the markers involved in apoptosis like caspase-3, caspase-9, Bax:Bcl-2 ratio, and cleaved PARP. 18β-GA also downregulated the expression of inflammatory proteins such as NF-κB, iNOS, and COX-2. Keeping these data into consideration, our results suggest that 18β-GA may be used as a chemopreventive agent in liver cancer.
Collapse
|
19
|
Akhmedov AT, Rybin V, Marín-García J. Mitochondrial oxidative metabolism and uncoupling proteins in the failing heart. Heart Fail Rev 2015; 20:227-49. [PMID: 25192828 DOI: 10.1007/s10741-014-9457-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite significant progress in cardiovascular medicine, myocardial ischemia and infarction, progressing eventually to the final end point heart failure (HF), remain the leading cause of morbidity and mortality in the USA. HF is a complex syndrome that results from any structural or functional impairment in ventricular filling or blood ejection. Ultimately, the heart's inability to supply the body's tissues with enough blood may lead to death. Mechanistically, the hallmarks of the failing heart include abnormal energy metabolism, increased production of reactive oxygen species (ROS) and defects in excitation-contraction coupling. HF is a highly dynamic pathological process, and observed alterations in cardiac metabolism and function depend on the disease progression. In the early stages, cardiac remodeling characterized by normal or slightly increased fatty acid (FA) oxidation plays a compensatory, cardioprotective role. However, upon progression of HF, FA oxidation and mitochondrial oxidative activity are decreased, resulting in a significant drop in cardiac ATP levels. In HF, as a compensatory response to decreased oxidative metabolism, glucose uptake and glycolysis are upregulated, but this upregulation is not sufficient to compensate for a drop in ATP production. Elevated mitochondrial ROS generation and ROS-mediated damage, when they overwhelm the cellular antioxidant defense system, induce heart injury and contribute to the progression of HF. Mitochondrial uncoupling proteins (UCPs), which promote proton leak across the inner mitochondrial membrane, have emerged as essential regulators of mitochondrial membrane potential, respiratory activity and ROS generation. Although the physiological role of UCP2 and UCP3, expressed in the heart, has not been clearly established, increasing evidence suggests that these proteins by promoting mild uncoupling could reduce mitochondrial ROS generation and cardiomyocyte apoptosis and ameliorate thereby myocardial function. Further investigation on the alterations in cardiac UCP activity and regulation will advance our understanding of their physiological roles in the healthy and diseased heart and also may facilitate the development of novel and more efficient therapies.
Collapse
Affiliation(s)
- Alexander T Akhmedov
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Avenue, Highland Park, NJ, 08904, USA
| | | | | |
Collapse
|
20
|
Stefano GB, Mantione KJ, Capellan L, Casares FM, Challenger S, Ramin R, Samuel JM, Snyder C, Kream RM. Morphine stimulates nitric oxide release in human mitochondria. J Bioenerg Biomembr 2015; 47:409-17. [PMID: 26350413 DOI: 10.1007/s10863-015-9626-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/03/2015] [Indexed: 10/23/2022]
Abstract
The expression of morphine by plants, invertebrate, and vertebrate cells and organ systems, strongly indicates a high level of evolutionary conservation of morphine and related morphinan alkaloids as required for life. The prototype catecholamine, dopamine, serves as an essential chemical intermediate in morphine biosynthesis, both in plants and animals. We surmise that, before the emergence of specialized plant and animal cells/organ systems, primordial multi-potential cell types required selective mechanisms to limit their responsiveness to environmental cues. Accordingly, cellular systems that emerged with the potential for recruitment of the free radical gas nitric oxide (NO) as a multi-faceted autocrine/paracrine signaling molecule, were provided with extremely positive evolutionary advantages. Endogenous morphinergic signaling, in concert with NO-coupled signaling systems, has evolved as an autocrine/paracrine regulator of metabolic homeostasis, energy metabolism, mitochondrial respiration and energy production. Basic physiological processes involving morphinergic/NO-coupled regulation of mitochondrial function, with special emphasis on the cardiovascular system, are critical to all organismic survival. Key to this concept may be the phenomenon of mitochondrial enslavement in eukaryotic evolution via endogenous morphine.
Collapse
Affiliation(s)
- George B Stefano
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA.
| | - Kirk J Mantione
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Lismary Capellan
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Federico M Casares
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Sean Challenger
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Rohina Ramin
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Joshua M Samuel
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Christopher Snyder
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| | - Richard M Kream
- MitoGenetics Research Institute, MitoGenetics LLC, 3 Bioscience Park Drive, Suite 307, Farmingdale, NY, 11735, USA
| |
Collapse
|
21
|
Basini G, Grasselli F. Nitric oxide in follicle development and oocyte competence. Reproduction 2015; 150:R1-9. [DOI: 10.1530/rep-14-0524] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/19/2015] [Indexed: 12/14/2022]
Abstract
Apart from its well-known role in regulating endothelial function, in mammals, nitric oxide (NO) is an important signaling molecule involved in many processes, regulating different biological functions. It has been demonstrated that NO plays a role in the physiology of the reproductive system, where it acts in controlling the activity of reproductive organs in both sexes. In the female of several animal species, experimental data suggest the presence of an intraovarian NO-generating system, which could be involved in the control of follicular development. The role of NO in regulating follicular atresia by apoptosis is still controversial, as a dual action depending mostly on its concentration has been documented. NO also displays positive effects on follicle development and selection related to angiogenic events and it could also play a modulatory role in steroidogenesis in ovarian cells. Both in monovulatory and poliovulatory species, the increase in PGE2production induced by NO via a stimulatory effect on COX-2 activity appears to be a common ovulatory mechanism. Considerable evidence also exists to support an involvement of the NO/NO synthase system in the control of meiotic maturation of cumulus–oocyte complexes.
Collapse
|
22
|
Vera S, Martínez R, Gormaz JG, Gajardo A, Galleguillos F, Rodrigo R. Novel relationships between oxidative stress and angiogenesis-related factors in sepsis: New biomarkers and therapies. Ann Med 2015; 47:289-300. [PMID: 25998489 DOI: 10.3109/07853890.2015.1029967] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Sepsis is a systemic uncontrolled inflammatory response in the presence of an infection. It remains a major cause of morbidity and mortality in hospitalized patients. According to its severity, sepsis can progress to three different states: severe sepsis, septic shock, and multiple organ dysfunction syndrome, related to organ dysfunction and/or tissue hypoperfusion. Different processes underlie its pathophysiology; among them are oxidative stress, endothelial and mitochondrial dysfunction, and angiogenesis-related factors. However, no studies have integrated these elements in sepsis. The main difficulty in sepsis is its diagnosis. Currently, the potential of inflammatory biomarkers in septic patients remains weak. In this context, the research into new biomarkers is essential to aid with sepsis diagnosis and prognostication. Furthermore, even though the current management of severe forms of sepsis has been effective, morbimortality remains elevated. Therefore, it is essential to explore alternative approaches to therapy development. The aim of this review is to present an update of evidence supporting the role of oxidative stress and angiogenesis-related factors in the pathophysiology of the different forms of sepsis. It proposes a novel convergence between both elements in their role in the disease, and it will cover their utility as new diagnostic tools, predictors of outcome, and as novel therapeutic targets.
Collapse
Affiliation(s)
- Sergio Vera
- Laboratory of Oxidative Stress and Nephrotoxicity, Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | | | | | | | | | | |
Collapse
|
23
|
Bak DW, Weerapana E. Cysteine-mediated redox signalling in the mitochondria. MOLECULAR BIOSYSTEMS 2015; 11:678-97. [DOI: 10.1039/c4mb00571f] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review represents a novel look at the many sources, cysteine targets, and signaling processes of ROS in the mitochondria.
Collapse
Affiliation(s)
- D. W. Bak
- Department of Chemistry
- Merkert Chemistry Center
- Boston College
- Massachusetts 02467
- USA
| | - E. Weerapana
- Department of Chemistry
- Merkert Chemistry Center
- Boston College
- Massachusetts 02467
- USA
| |
Collapse
|
24
|
|
25
|
Lee CC, Hsieh HJ, Hsieh CH, Hwang DF. Spine venom of crown-of-thorns starfish (Acanthaster planci) induces antiproliferation and apoptosis of human melanoma cells (A375.S2). Toxicon 2014; 91:126-34. [PMID: 25159188 DOI: 10.1016/j.toxicon.2014.08.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 08/04/2014] [Accepted: 08/14/2014] [Indexed: 01/08/2023]
Abstract
The crown-of-thorns starfish (Acanthaster planci) is a venomous starfish. In this study, the extraction of A. planci spine venom (ASV) was performed by phosphate saline buffer, followed by assaying the cytotoxicity on human normal and tumor cells. It was found that human melanoma cells (A375.S2) were the most sensitive to the ASV solution. The cells, after incubation with ASV, significantly appeared to decrease cell viability and increase lactate dehydrogenase (LDH) release with a dose-dependent relationship. The extract of spine promoted loss of mitochondrial membrane potential (ΔΨm) and induced inter-nucleosomal DNA fragmentation in human melanoma cells. The cells exhibited apoptosis by using propidium iodide (PI) staining of DNA fragmentation; it was then determined by flow cytometry (sub-G1 peak). The molecular cytotoxicity of ASV was tested through evaluation of the apoptosis/necrosis ratio by double staining with annexin V and PI assay. The A. planci spine venom showed significant antiproliferation. The human melanoma cells revealed apoptosis at low dose (1.25 μg/ml), and necrosis occurred at high dose (5 μg/ml).
Collapse
Affiliation(s)
- Chi-Chiu Lee
- Department of Food Science and Center of Excellence for the Ocean, National Taiwan Ocean University, 2 Pei-Ning Road, Keelung 202, Taiwan, ROC
| | - Hernyi Justin Hsieh
- Penghu Marine Biology Research Center, Fishery Research Institute, Council of Agriculture, Magong, Penghu 880, Taiwan, ROC
| | - Cheng-Hong Hsieh
- Department of Health and Nutrition Biotechnology, Asia University, 500, Lioufeng Road, Wufeng, Taichung 413, Taiwan, ROC
| | - Deng-Fwu Hwang
- Department of Food Science and Center of Excellence for the Ocean, National Taiwan Ocean University, 2 Pei-Ning Road, Keelung 202, Taiwan, ROC; Department of Health and Nutrition Biotechnology, Asia University, 500, Lioufeng Road, Wufeng, Taichung 413, Taiwan, ROC.
| |
Collapse
|
26
|
Joshi-Barr S, de Gracia Lux C, Mahmoud E, Almutairi A. Exploiting oxidative microenvironments in the body as triggers for drug delivery systems. Antioxid Redox Signal 2014; 21:730-54. [PMID: 24328819 PMCID: PMC4098119 DOI: 10.1089/ars.2013.5754] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SIGNIFICANCE Reactive oxygen species and reactive nitrogen species (ROS/RNS) play an important role in cell signaling pathways. However, the increased production of these species may disrupt cellular homeostasis, giving rise to pathological conditions. Biomaterials that are responsive to ROS/RNS can be strategically used to specifically release therapeutics and diagnostic agents to regions undergoing oxidative stress. RECENT ADVANCES Many nanocarriers intended to exploit redox micro-environments as triggers for drug release, summarized and compared in this review, have recently been developed. We describe these carriers' chemical structures, strategies for payload protection and oxidation-selective release, and ROS/RNS sensitivity as tested in initial studies. CRITICAL ISSUES ROS/RNS are unstable, so reliable measures of their concentrations in various conditions are scarce. Combined with the dearth of materials shown to respond to physiologically relevant levels of ROS/RNS, evaluations of their true sensitivity are difficult. FUTURE DIRECTIONS Oxidation-responsive nanocarriers developed thus far show tremendous potential for applicability in vivo; however, the sensitivity of these chemistries needs to be fine tuned to enable responses to physiological levels of ROS and RNS.
Collapse
Affiliation(s)
- Shivanjali Joshi-Barr
- 1 Skaggs School of Pharmacy and Pharmaceutical Sciences, Laboratory of Bioresponsive Materials, University of California , San Diego, San Diego, California
| | | | | | | |
Collapse
|
27
|
Sansbury BE, Hill BG. Regulation of obesity and insulin resistance by nitric oxide. Free Radic Biol Med 2014; 73:383-99. [PMID: 24878261 PMCID: PMC4112002 DOI: 10.1016/j.freeradbiomed.2014.05.016] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/16/2014] [Accepted: 05/17/2014] [Indexed: 02/07/2023]
Abstract
Obesity is a risk factor for developing type 2 diabetes and cardiovascular disease and has quickly become a worldwide pandemic with few tangible and safe treatment options. Although it is generally accepted that the primary cause of obesity is energy imbalance, i.e., the calories consumed are greater than are utilized, understanding how caloric balance is regulated has proven a challenge. Many "distal" causes of obesity, such as the structural environment, occupation, and social influences, are exceedingly difficult to change or manipulate. Hence, molecular processes and pathways more proximal to the origins of obesity-those that directly regulate energy metabolism or caloric intake-seem to be more feasible targets for therapy. In particular, nitric oxide (NO) is emerging as a central regulator of energy metabolism and body composition. NO bioavailability is decreased in animal models of diet-induced obesity and in obese and insulin-resistant patients, and increasing NO output has remarkable effects on obesity and insulin resistance. This review discusses the role of NO in regulating adiposity and insulin sensitivity and places its modes of action into context with the known causes and consequences of metabolic disease.
Collapse
Affiliation(s)
- Brian E Sansbury
- Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bradford G Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
28
|
Wang F, Guo X, Shen X, Kream RM, Mantione KJ, Stefano GB. Vascular dysfunction associated with type 2 diabetes and Alzheimer's disease: a potential etiological linkage. Med Sci Monit Basic Res 2014; 20:118-29. [PMID: 25082505 PMCID: PMC4138067 DOI: 10.12659/msmbr.891278] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The endothelium performs a crucial role in maintaining vascular integrity leading to whole organ metabolic homeostasis. Endothelial dysfunction represents a key etiological factor leading to moderate to severe vasculopathies observed in both Type 2 diabetic and Alzheimer’s Disease (AD) patients. Accordingly, evidence-based epidemiological factors support a compelling hypothesis stating that metabolic rundown encountered in Type 2 diabetes engenders severe cerebral vascular insufficiencies that are causally linked to long term neural degenerative processes in AD. Of mechanistic importance, Type 2 diabetes engenders an immunologically mediated chronic pro-inflammatory state involving interactive deleterious effects of leukocyte-derived cytokines and endothelial-derived chemotactic agents leading to vascular and whole organ dysfunction. The long term negative consequences of vascular pro-inflammatory processes on the integrity of CNS basal forebrain neuronal populations mediating complex cognitive functions establish a striking temporal comorbidity of AD with Type 2 diabetes. Extensive biomedical evidence supports the pivotal multi-functional role of constitutive nitric oxide (NO) production and release as a critical vasodilatory, anti-inflammatory, and anti-oxidant, mechanism within the vascular endothelium. Within this context, we currently review the functional contributions of dysregulated endothelial NO expression to the etiology and persistence of Type 2 diabetes-related and co morbid AD-related vasculopathies. Additionally, we provide up-to-date perspectives on critical areas of AD research with special reference to common NO-related etiological factors linking Type 2 diabetes to the pathogenesis of AD.
Collapse
Affiliation(s)
- Fuzhou Wang
- Department of Anesthesiology and Critical Care Medicine, Nanjing Maternit and Child Health Care Hospital, Nanjing Medical University, Nanjing, China (mainland)
| | - Xirong Guo
- Institutes of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China (mainland)
| | - Xiaofeng Shen
- Department of Anesthesiology and Critical Care Medicine, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China (mainland)
| | - Richard M Kream
- Neuroscience Research Institute, State University of New York - College at Old Westbury, Old Westbury, USA
| | - Kirk J Mantione
- Neuroscience Research Institute, State University of New York - College at Old Westbury, Old Westbury, USA
| | - George B Stefano
- Neuroscience Research Institute, State University of New York - College at Old Westbury, Old Westbury, USA
| |
Collapse
|
29
|
Lee CC, Hsieh HJ, Hwang DF. Cytotoxic and apoptotic activities of the plancitoxin I from the venom of crown-of-thorns starfish (Acanthaster planci) on A375.S2 cells. J Appl Toxicol 2014; 35:407-17. [PMID: 25047904 DOI: 10.1002/jat.3034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/16/2014] [Accepted: 05/16/2014] [Indexed: 02/06/2023]
Abstract
This study reports on a cytotoxic toxin derived from the venom of the crown-of-thorns starfish Acanthaster planci (CAV). The protein toxin was isolated through both ion-exchange and gel-filtration chromatography, and characterized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and mass spectrum analyzes. The CAV was identified as plancitoxin I protein. The mechanistic role of the CAV toxin was explored in human malignant melanoma A375.S2 cell death. The results indicated that after incubation with CAV toxin, cells significantly decreased in A375.S2 cell viability and increased in the lactate dehydrogenase (LDH) level in a dose-dependent manner. The assays indicated that CAV toxin promoted reactive oxygen species (ROS) production, induced nitric oxide (NO) formation, lost mitochondrial membrane potential (ΔΨm) and induced inter-nucleosomal DNA fragmentation in A375.S2 cells. The molecular cytotoxicity of the CAV toxin was tested through evaluation of the apoptosis/necrosis ratio by double staining with annexin V-FITC and a propidium iodide (PI) assay. The results suggested that CAV toxin induced a cytotoxic effect in A375.S2 cells via the apoptotic procedure, and may be associated with the regulation of the p38 pathways.
Collapse
Affiliation(s)
- Chi-Chiu Lee
- Department of Food Science and Center of Excellence for Marine Bioenvironment and Biotechnology, National Taiwan Ocean University, 2 Pei-Ning Road, Keelung, 202, Taiwan, ROC
| | | | | |
Collapse
|
30
|
De Nicola AF, Gonzalez Deniselle MC, Garay L, Meyer M, Gargiulo-Monachelli G, Guennoun R, Schumacher M, Carreras MC, Poderoso JJ. Progesterone protective effects in neurodegeneration and neuroinflammation. J Neuroendocrinol 2013; 25:1095-103. [PMID: 23639063 DOI: 10.1111/jne.12043] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 04/12/2013] [Accepted: 04/26/2013] [Indexed: 01/03/2023]
Abstract
Progesterone is a neuroprotective, promyelinating and anti-inflammatory factor for the nervous system. Here, we review the effects of progesterone in models of motoneurone degeneration and neuroinflammation. In neurodegeneration of the Wobbler mouse, a subset of spinal cord motoneurones showed increased activity of nitric oxide synthase (NOS), increased intramitochondrial NOS, decreased activity of respiratory chain complexes, and decreased activity and protein expression of Mn-superoxide dismutase type 2 (MnSOD2). Clinically, Wobblers suffered several degrees of motor impairment. Progesterone treatment restored the expression of neuronal markers, decreased the activity of NOS and enhanced complex I respiratory activity and MnSOD2. Long-term treatment with progesterone increased muscle strength, biceps weight and survival. Collectively, these data suggest that progesterone prevented neurodegeneration. To study the effects of progesterone in neuroinflammation, we employed mice with experimental autoimmune encephalomyelitis (EAE). EAE mice spinal cord showed increased mRNA levels of the inflammatory mediators tumour necrosis factor (TNF)α and its receptor TNFR1, the microglial marker CD11b, inducible NOS and the toll-like receptor 4. Progesterone pretreatment of EAE mice blocked the proinflammatory mediators, decreased Iba1+ microglial cells and attenuated clinical signs of EAE. Therefore, reactive glial cells became targets of progesterone anti-inflammatory effects. These results represent a starting point for testing the usefulness of neuroactive steroids in neurological disorders.
Collapse
Affiliation(s)
- A F De Nicola
- Department of Human Biochemistry, Faculty of Medicine, Instituto de Biologia y Medicina Experimental, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Based on mosaic theory, hypertension is a multifactorial disorder that develops because of genetic, environmental, anatomical, adaptive neural, endocrine, humoral, and hemodynamic factors. It has been recently proposed that oxidative stress may contribute to all of these factors and production of reactive oxygen species (ROS) play an important role in the development of hypertension. Previous studies focusing on the role of vascular NADPH oxidases provided strong support of this concept. Although mitochondria represent one of the most significant sources of cellular ROS generation, the regulation of mitochondrial ROS generation in the cardiovascular system and its pathophysiological role in hypertension are much less understood. In this review, the role of mitochondrial oxidative stress in the pathophysiology of hypertension and cross talk between angiotensin II signaling, pathways involved in mechanotransduction, NADPH oxidases, and mitochondria-derived ROS are considered. The possible benefits of therapeutic strategies that have the potential to attenuate mitochondrial oxidative stress for the prevention/treatment of hypertension are also discussed.
Collapse
Affiliation(s)
- Sergey I Dikalov
- Division of Clinical Pharmacology, Free Radicals in Medicine Core, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | | |
Collapse
|
32
|
Campos JC, Gomes KMS, Ferreira JCB. Impact of exercise training on redox signaling in cardiovascular diseases. Food Chem Toxicol 2013; 62:107-19. [PMID: 23978413 DOI: 10.1016/j.fct.2013.08.035] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/05/2013] [Accepted: 08/18/2013] [Indexed: 02/07/2023]
Abstract
Reactive oxygen and nitrogen species regulate a wide array of signaling pathways that governs cardiovascular physiology. However, oxidant stress resulting from disrupted redox signaling has an adverse impact on the pathogenesis and progression of cardiovascular diseases. In this review, we address how redox signaling and oxidant stress affect the pathophysiology of cardiovascular diseases such as ischemia-reperfusion injury, hypertension and heart failure. We also summarize the benefits of exercise training in tackling the hyperactivation of cellular oxidases and mitochondrial dysfunction seen in cardiovascular diseases.
Collapse
Affiliation(s)
- Juliane C Campos
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | |
Collapse
|
33
|
Noriega-Cisneros R, Cortés-Rojo C, Manzo-Avalos S, Clemente-Guerrero M, Calderón-Cortés E, Salgado-Garciglia R, Montoya-Pérez R, Boldogh I, Saavedra-Molina A. Mitochondrial response to oxidative and nitrosative stress in early stages of diabetes. Mitochondrion 2013; 13:835-40. [PMID: 23751425 DOI: 10.1016/j.mito.2013.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 05/04/2013] [Accepted: 05/30/2013] [Indexed: 12/23/2022]
Abstract
Diabetes mellitus (DM) is associated with increased production of reactive oxygen and nitrogen species; consequently, an increase in lipid peroxidation and a decrease in antioxidants resulting in mitochondrial dysfunction. Using a rat model of DM induced by streptozotocin, we show the opposite: an increase in NO levels, S-nitrosylation, aconitase activity, and total glutathione and a decrease in lipid peroxidation at early stages of diabetes. These data imply that the decrease in lipid peroxidation is a vital early response to hyperglycemia to prevent escalation of ROS generation in mitochondria. These results also suggest a need for novel therapeutic targets to prevent the neurological consequences of diabetes.
Collapse
Affiliation(s)
- Ruth Noriega-Cisneros
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mich., Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Common polymorphisms in nitric oxide synthase (NOS) genes influence quality of aging and longevity in humans. Biogerontology 2013; 14:177-86. [PMID: 23572278 DOI: 10.1007/s10522-013-9421-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/03/2013] [Indexed: 12/22/2022]
Abstract
Nitric oxide (NO) triggers multiple signal transduction pathways and contributes to the control of numerous cellular functions. Previous studies have shown in model organisms that the alteration of NO production has important effects on aging and lifespan. We studied in a large sample (763 subjects, age range 19-107 years) the variability of the three human genes (NOS1, -2, -3) coding for the three isoforms of the NADPH-dependent enzymes named NO synthases (NOS) which are responsible of NO synthesis. We have then verified if the variability of these genes is associated with longevity, and with a number of geriatric parameters. We found that gene variation of NOS1 and NOS2 was associated with longevity. In addition NOS1 rs1879417 was also found to be associated with a lower cognitive performance, while NOS2 rs2297518 polymorphism showed to be associated with physical performance. Moreover, SNPs in the NOS1 and NOS3 genes were respectively associated with the presence of depression symptoms and disability, two of the main factors affecting quality of life in older individuals. On the whole, our study shows that genetic variability of NOS genes has an effect on common age related phenotypes and longevity in humans as well as previously reported for model organisms.
Collapse
|
35
|
Bolisetty S, Jaimes EA. Mitochondria and reactive oxygen species: physiology and pathophysiology. Int J Mol Sci 2013; 14:6306-44. [PMID: 23528859 PMCID: PMC3634422 DOI: 10.3390/ijms14036306] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/08/2013] [Accepted: 03/11/2013] [Indexed: 02/06/2023] Open
Abstract
The air that we breathe contains nearly 21% oxygen, most of which is utilized by mitochondria during respiration. While we cannot live without it, it was perceived as a bane to aerobic organisms due to the generation of reactive oxygen and nitrogen metabolites by mitochondria and other cellular compartments. However, this dogma was challenged when these species were demonstrated to modulate cellular responses through altering signaling pathways. In fact, since this discovery of a dichotomous role of reactive species in immune function and signal transduction, research in this field grew at an exponential pace and the pursuit for mechanisms involved began. Due to a significant number of review articles present on the reactive species mediated cell death, we have focused on emerging novel pathways such as autophagy, signaling and maintenance of the mitochondrial network. Despite its role in several processes, increased reactive species generation has been associated with the origin and pathogenesis of a plethora of diseases. While it is tempting to speculate that anti-oxidant therapy would protect against these disorders, growing evidence suggests that this may not be true. This further supports our belief that these reactive species play a fundamental role in maintenance of cellular and tissue homeostasis.
Collapse
Affiliation(s)
- Subhashini Bolisetty
- Nephrology Division, University of Alabama at Birmingham, Birmingham, AL 35294, USA; E-Mail:
| | - Edgar A. Jaimes
- Nephrology Division, University of Alabama at Birmingham, Birmingham, AL 35294, USA; E-Mail:
- Veterans Affairs Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
36
|
Shimizu S, Ishibashi M, Kumagai S, Wajima T, Hiroi T, Kurihara T, Ishii M, Kiuchi Y. Decreased cardiac mitochondrial tetrahydrobiopterin in a rat model of pressure overload. Int J Mol Med 2013; 31:589-96. [PMID: 23313998 DOI: 10.3892/ijmm.2013.1236] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 12/19/2012] [Indexed: 11/05/2022] Open
Abstract
Sustained cardiac pressure overload induces mitochondrial dysfunction and apoptosis of cardiomyocytes leading to pathological cardiac hypertrophy and dysfunction. Mitochondrial nitric oxide synthase (NOS) appears to cause uncoupling, which produces reactive oxygen species (ROS) instead of nitric oxide (NO), by a decrease in the cofactor tetrahydrobiopterin (BH4). This study focused on examining the changes in mitochondrial BH4 levels during cardiac pressure overload. Chronic cardiac pressure overload was generated by abdominal aortic banding in rats. Levels of BH4 and its oxidized form were measured in the mitochondria isolated from the left ventricle (LV) and the post-mitochondrial supernatants. Chronic aortic banding increased blood pressure, and induced cardiac hypertrophy and fibrosis. Notably, the BH4 levels were decreased while its oxidized forms were increased in LV mitochondria, but not in the post-mitochondrial supernatants containing the cytosol and microsome. Anti-neuronal NOS antibody-sensitive protein was detected in the cardiac mitochondria. Moreover, continuous administration of BH4 to rats with pressure overload increased mitochondrial BH4 levels and reduced cardiac fibrosis and matrix metallopeptidase activity, but not cardiac hypertrophy. These findings show the possibility that NOS uncoupling by decreased cardiac mitochondrial BH4 levels is implicated, at least in part, in the development of cardiac fibrosis, leading to cardiac dysfunction induced by pressure overload.
Collapse
Affiliation(s)
- Shunichi Shimizu
- Department of Pathophysiology, Showa University School of Pharmacy, Tokyo 142-8555, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Penna C, Perrelli MG, Pagliaro P. Mitochondrial pathways, permeability transition pore, and redox signaling in cardioprotection: therapeutic implications. Antioxid Redox Signal 2013; 18:556-99. [PMID: 22668069 DOI: 10.1089/ars.2011.4459] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Reperfusion therapy is the indispensable treatment of acute myocardial infarction (AMI) and must be applied as soon as possible to attenuate the ischemic insult. However, reperfusion is responsible for additional myocardial damage likely involving opening of the mitochondrial permeability transition pore (mPTP). A great part of reperfusion injury occurs during the first minute of reperfusion. The prolonged opening of mPTP is considered one of the endpoints of the cascade to myocardial damage, causing loss of cardiomyocyte function and viability. Opening of mPTP and the consequent oxidative stress due to reactive oxygen and nitrogen species (ROS/RNS) are considered among the major mechanisms of mitochondrial and myocardial dysfunction. Kinases and mitochondrial components constitute an intricate network of signaling molecules and mitochondrial proteins, which interact in response to stressors. Cardioprotective pathways are activated by stimuli such as preconditioning and postconditioning (PostC), obtained with brief intermittent ischemia or with pharmacological agents, which drastically reduce the lethal ischemia/reperfusion injury. The protective pathways converging on mitochondria may preserve their function. Protection involves kinases, adenosine triphosphate-dependent potassium channels, ROS signaling, and the mPTP modulation. Some clinical studies using ischemic PostC during angioplasty support its protective effects, and an interesting alternative is pharmacological PostC. In fact, the mPTP desensitizer, cyclosporine A, has been shown to induce appreciable protections in AMI patients. Several factors and comorbidities that might interfere with cardioprotective signaling are considered. Hence, treatments adapted to the characteristics of the patient (i.e., phenotype oriented) might be feasible in the future.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | | | | |
Collapse
|
38
|
Nitric oxide in skeletal muscle: role on mitochondrial biogenesis and function. Int J Mol Sci 2012; 13:17160-84. [PMID: 23242154 PMCID: PMC3546744 DOI: 10.3390/ijms131217160] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/04/2012] [Accepted: 12/05/2012] [Indexed: 01/18/2023] Open
Abstract
Nitric oxide (NO) has been implicated in several cellular processes as a signaling molecule and also as a source of reactive nitrogen species (RNS). NO is produced by three isoenzymes called nitric oxide synthases (NOS), all present in skeletal muscle. While neuronal NOS (nNOS) and endothelial NOS (eNOS) are isoforms constitutively expressed, inducible NOS (iNOS) is mainly expressed during inflammatory responses. Recent studies have demonstrated that NO is also involved in the mitochondrial biogenesis pathway, having PGC-1α as the main signaling molecule. Increased NO synthesis has been demonstrated in the sarcolemma of skeletal muscle fiber and NO can also reversibly inhibit cytochrome c oxidase (Complex IV of the respiratory chain). Investigation on cultured skeletal myotubes treated with NO donors, NO precursors or NOS inhibitors have also showed a bimodal effect of NO that depends on the concentration used. The present review will discuss the new insights on NO roles on mitochondrial biogenesis and function in skeletal muscle. We will also focus on potential therapeutic strategies based on NO precursors or analogs to treat patients with myopathies and mitochondrial deficiency.
Collapse
|
39
|
Dey R, Kemp K, Gray E, Rice C, Scolding N, Wilkins A. Human mesenchymal stem cells increase anti-oxidant defences in cells derived from patients with Friedreich's ataxia. CEREBELLUM (LONDON, ENGLAND) 2012; 11:861-71. [PMID: 22826109 DOI: 10.1007/s12311-012-0406-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Friedreich's ataxia (FRDA) is a progressive neurodegenerative disorder which is, at present, incurable. Oxidative damage and inhibition of mitochondrial function are key determinants of cellular damage in FRDA, since there is greater sensitivity to oxidative stress in cells with frataxin deficiency. In addition, frataxin-deficient cells have an impaired ability to recruit antioxidant defences against endogenous oxidative stress. We have recently shown that factors derived from bone marrow-derived mesenchymal stem cells (MSCs) increase hydrogen peroxide scavenging enzymes and offer protection against hydrogen peroxide-mediated injury in cells derived from patients with FRDA. Here we extend these studies and have performed a series of experiments showing that expression of superoxide dismutase (1 and 2) enzymes is reduced in FRDA cells but can be restored by treatment with conditioned medium from human MSCs. Furthermore, we have demonstrated that exposure to factors secreted by MSCs increases resistance to nitric oxide-induced oxidative stress in FRDA fibroblasts through, at least in part, restoring the expression of the superoxide dismuting enzymes and via modulation of PI(3) kinase/Akt pathways. These findings suggest that MSCs secrete factors that improve the cellular homeostasis of cells derived from FRDA patients and provide suitable support for their enhanced survival. This study further suggests the potential therapeutic use of MSCs in patients with FRDA.
Collapse
Affiliation(s)
- Rimi Dey
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol, UK
| | | | | | | | | | | |
Collapse
|
40
|
Olson KR, Donald JA, Dombkowski RA, Perry SF. Evolutionary and comparative aspects of nitric oxide, carbon monoxide and hydrogen sulfide. Respir Physiol Neurobiol 2012; 184:117-29. [DOI: 10.1016/j.resp.2012.04.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 04/09/2012] [Accepted: 04/15/2012] [Indexed: 12/13/2022]
|
41
|
Zhuravliova E, Barbakadze T, Jojua N, Zaalishvili E, Shanshiashvili L, Natsvlishvili N, Kalandadze I, Narmania N, Chogovadze I, Mikeladze D. Synaptic and non-synaptic mitochondria in hippocampus of adult rats differ in their sensitivity to hypothyroidism. Cell Mol Neurobiol 2012; 32:1311-21. [PMID: 22706894 DOI: 10.1007/s10571-012-9857-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 05/31/2012] [Indexed: 11/28/2022]
Abstract
Hypothyroidism in humans provokes various neuropsychiatric disorders, movement, and cognitive abnormalities that may greatly depend on the mitochondrial energy metabolism. Brain cells contain at least two major populations of mitochondria that include the non-synaptic mitochondria, which originate from neuronal and glial cell bodies (CM), and the synaptic (SM) mitochondria, which primarily originate from the nerve terminals. Several parameters of oxidative stress and other parameters in SM and CM fractions of hippocampus of adult rats were compared among euthyroid (control), hypothyroid (methimazol-treated), and thyroxine (T4)-treated hypothyroid states. nNOS translocation to CM was observed with concomitant increase of mtNOS's activity in hypothyroid rats. In parallel, oxidation of cytochrome c oxidase and production of peroxides with substrates of complex I (glutamate + malate) were enhanced in CM, whereas the activity of aconitase and mitochondrial membrane potential (ΔΨm) were decreased. Furthermore, the elevation of mitochondrial hexokinase activity in CM was also found. No differences in these parameters between control and hypothyroid animals were observed in SM. However, in contrast to CM, hypothyroidism increases the level of pro-apoptotic K-Ras and Bad in SM. Our results suggest that hypothyroidism induces moderate and reversible oxidative/nitrosative stress in hippocampal CM, leading to the compensatory elevation of hexokinase activity and aerobic glycolysis. Such adaptive activation in glycolytic metabolism does not occur in SM, suggesting that synaptic mitochondria differ in their sensitivity to the energetic disturbance in hypothyroid conditions.
Collapse
Affiliation(s)
- E Zhuravliova
- I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Long J, Aksenov V, Rollo CD, Liu J. A complex dietary supplement modulates nitrative stress in normal mice and in a new mouse model of nitrative stress and cognitive aging. Mech Ageing Dev 2012; 133:523-9. [DOI: 10.1016/j.mad.2012.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 03/17/2012] [Accepted: 04/05/2012] [Indexed: 01/07/2023]
|
43
|
Zádori D, Klivényi P, Szalárdy L, Fülöp F, Toldi J, Vécsei L. Mitochondrial disturbances, excitotoxicity, neuroinflammation and kynurenines: novel therapeutic strategies for neurodegenerative disorders. J Neurol Sci 2012; 322:187-91. [PMID: 22749004 DOI: 10.1016/j.jns.2012.06.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 06/08/2012] [Indexed: 12/25/2022]
Abstract
A mitochondrial dysfunction causes an abatement in ATP production, the induction of oxidative damage and the propagation of cell death pathways. It is additionally closely related to both glutamate excitotoxicity and neuroinflammation. All of these interconnected aspects of a cellular dysfunction are involved in the pathogenesis of numerous neurological disorders, including those with an acute (e.g. ischemic stroke) or a chronic (e.g. Huntington's disease) onset. Both acute and chronic neurodegenerative disorders have been demonstrated to involve multiple imbalances of the kynurenine pathway metabolism in the pathogenesis of the disease. As regards neuroactive compounds featuring in the pathway, quinolinic acid is a specific agonist of N-methyl-d-aspartate receptors, and a potent neurotoxin with additional and marked free radical-producing and lipid peroxidation-inducing properties. The toxic effects of 3-hydroxy-L-kynurenine are mediated by free radicals. Besides the possibility of increasing brain kynurenic acid concentrations, L-kynurenine may have vasoactive properties, too. Kynurenic acid has proven to be neuroprotective in several experimental settings, but in consequence of its pharmacokinetic properties it is not applicable as systemic administration in human cases. The aim of this short review is to emphasize the common features of cerebral ischemia and Huntington's disease and to highlight therapeutic strategies targeting the kynurenine pathway.
Collapse
Affiliation(s)
- Dénes Zádori
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary.
| | | | | | | | | | | |
Collapse
|
44
|
Dai DF, Chen T, Johnson SC, Szeto H, Rabinovitch PS. Cardiac aging: from molecular mechanisms to significance in human health and disease. Antioxid Redox Signal 2012; 16:1492-526. [PMID: 22229339 PMCID: PMC3329953 DOI: 10.1089/ars.2011.4179] [Citation(s) in RCA: 224] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVDs) are the major causes of death in the western world. The incidence of cardiovascular disease as well as the rate of cardiovascular mortality and morbidity increase exponentially in the elderly population, suggesting that age per se is a major risk factor of CVDs. The physiologic changes of human cardiac aging mainly include left ventricular hypertrophy, diastolic dysfunction, valvular degeneration, increased cardiac fibrosis, increased prevalence of atrial fibrillation, and decreased maximal exercise capacity. Many of these changes are closely recapitulated in animal models commonly used in an aging study, including rodents, flies, and monkeys. The application of genetically modified aged mice has provided direct evidence of several critical molecular mechanisms involved in cardiac aging, such as mitochondrial oxidative stress, insulin/insulin-like growth factor/PI3K pathway, adrenergic and renin angiotensin II signaling, and nutrient signaling pathways. This article also reviews the central role of mitochondrial oxidative stress in CVDs and the plausible mechanisms underlying the progression toward heart failure in the susceptible aging hearts. Finally, the understanding of the molecular mechanisms of cardiac aging may support the potential clinical application of several "anti-aging" strategies that treat CVDs and improve healthy cardiac aging.
Collapse
Affiliation(s)
- Dao-Fu Dai
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | | | | | | | | |
Collapse
|
45
|
Deniselle MCG, Carreras MC, Garay L, Gargiulo-Monachelli G, Meyer M, Poderoso JJ, De Nicola AF. Progesterone prevents mitochondrial dysfunction in the spinal cord of wobbler mice. J Neurochem 2012; 122:185-95. [DOI: 10.1111/j.1471-4159.2012.07753.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
Chen Z, Li Q, Sun Q, Chen H, Wang X, Li N, Yin M, Xie Y, Li H, Tang B. Simultaneous Determination of Reactive Oxygen and Nitrogen Species in Mitochondrial Compartments of Apoptotic HepG2 Cells and PC12 Cells Based On Microchip Electrophoresis–Laser-Induced Fluorescence. Anal Chem 2012; 84:4687-94. [DOI: 10.1021/ac300255n] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
47
|
Abstract
Old age is a major risk factor for cardiovascular diseases. Several lines of evidence in experimental animal models have indicated the central role of mitochondria both in lifespan determination and in cardiovascular aging. In this article we review the evidence supporting the role of mitochondrial oxidative stress, mitochondrial damage and biogenesis as well as the crosstalk between mitochondria and cellular signaling in cardiac and vascular aging. Intrinsic cardiac aging in the murine model closely recapitulates age-related cardiac changes in humans (left ventricular hypertrophy, fibrosis and diastolic dysfunction), while the phenotype of vascular aging include endothelial dysfunction, reduced vascular elasticity, and chronic vascular inflammation. Both cardiac and vascular aging involve neurohormonal signaling (eg, renin-angiotensin, adrenergic, insulin-IGF1 signaling) and cell-autonomous mechanisms. The potential therapeutic strategies to improve mitochondrial function in aging and cardiovascular diseases are also discussed, with a focus on mitochondrial-targeted antioxidants, calorie restriction, calorie restriction mimetics, and exercise training.
Collapse
Affiliation(s)
- Dao-Fu Dai
- Department of Pathology, University of Washington, Seattle, USA
| | | | | |
Collapse
|
48
|
Antico Arciuch VG, Elguero ME, Poderoso JJ, Carreras MC. Mitochondrial regulation of cell cycle and proliferation. Antioxid Redox Signal 2012; 16:1150-80. [PMID: 21967640 PMCID: PMC3315176 DOI: 10.1089/ars.2011.4085] [Citation(s) in RCA: 294] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 10/03/2011] [Accepted: 10/03/2011] [Indexed: 01/01/2023]
Abstract
Eukaryotic mitochondria resulted from symbiotic incorporation of α-proteobacteria into ancient archaea species. During evolution, mitochondria lost most of the prokaryotic bacterial genes and only conserved a small fraction including those encoding 13 proteins of the respiratory chain. In this process, many functions were transferred to the host cells, but mitochondria gained a central role in the regulation of cell proliferation and apoptosis, and in the modulation of metabolism; accordingly, defective organelles contribute to cell transformation and cancer, diabetes, and neurodegenerative diseases. Most cell and transcriptional effects of mitochondria depend on the modulation of respiratory rate and on the production of hydrogen peroxide released into the cytosol. The mitochondrial oxidative rate has to remain depressed for cell proliferation; even in the presence of O₂, energy is preferentially obtained from increased glycolysis (Warburg effect). In response to stress signals, traffic of pro- and antiapoptotic mitochondrial proteins in the intermembrane space (B-cell lymphoma-extra large, Bcl-2-associated death promoter, Bcl-2 associated X-protein and cytochrome c) is modulated by the redox condition determined by mitochondrial O₂ utilization and mitochondrial nitric oxide metabolism. In this article, we highlight the traffic of the different canonical signaling pathways to mitochondria and the contributions of organelles to redox regulation of kinases. Finally, we analyze the dynamics of the mitochondrial population in cell cycle and apoptosis.
Collapse
Affiliation(s)
| | - María Eugenia Elguero
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
| | - Juan José Poderoso
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- Department of Internal Medicine, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - María Cecilia Carreras
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
- Department of Clinical Biochemistry, INFIBIOC and School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
49
|
Du J, Teng RJ, Lawrence M, Guan T, Xu H, Ge Y, Shi Y. The protein partners of GTP cyclohydrolase I in rat organs. PLoS One 2012; 7:e33991. [PMID: 22479495 PMCID: PMC3313957 DOI: 10.1371/journal.pone.0033991] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 02/22/2012] [Indexed: 02/03/2023] Open
Abstract
Objective GTP cyclohydrolase I (GCH1) is the rate-limiting enzyme for tetrahydrobiopterin biosynthesis and has been shown to be a promising therapeutic target in ischemic heart disease, hypertension, atherosclerosis and diabetes. The endogenous GCH1-interacting partners have not been identified. Here, we determined endogenous GCH1-interacting proteins in rat. Methods and Results A pulldown and proteomics approach were used to identify GCH1 interacting proteins in rat liver, brain, heart and kidney. We demonstrated that GCH1 interacts with at least 17 proteins including GTP cyclohydrolase I feedback regulatory protein (GFRP) in rat liver by affinity purification followed by proteomics and validated six protein partners in liver, brain, heart and kidney by immunoblotting. GCH1 interacts with GFRP and very long-chain specific acyl-CoA dehydrogenase in the liver, tubulin beta-2A chain in the liver and brain, DnaJ homolog subfamily A member 1 and fatty aldehyde dehydrogenase in the liver, heart and kidney and eukaryotic translation initiation factor 3 subunit I (EIF3I) in all organs tested. Furthermore, GCH1 associates with mitochondrial proteins and GCH1 itself locates in mitochondria. Conclusion GCH1 interacts with proteins in an organ dependant manner and EIF3I might be a general regulator of GCH1. Our finding indicates GCH1 might have broader functions beyond tetrahydrobiopterin biosynthesis.
Collapse
Affiliation(s)
- Jianhai Du
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail: (JD); (YS)
| | - Ru-Jeng Teng
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Matt Lawrence
- Human Proteomics Program and Department of Physiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Tongju Guan
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Hao Xu
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ying Ge
- Human Proteomics Program and Department of Physiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Yang Shi
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Patient Centered Research, Aurora Health Care, Milwaukee, Wisconsin, United States of America
- * E-mail: (JD); (YS)
| |
Collapse
|
50
|
Radak Z, Naito H, Taylor AW, Goto S. Nitric oxide: Is it the cause of muscle soreness? Nitric Oxide 2012; 26:89-94. [DOI: 10.1016/j.niox.2011.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 12/14/2011] [Accepted: 12/21/2011] [Indexed: 11/25/2022]
|