101
|
Li K, You T, Zhao P, Luo Y, Zhang D, Wei H, Wang Y, Yang J, Guan X, Kuang Z. Structural basis for the regulation of inducible nitric oxide synthase by the SPRY domain-containing SOCS box protein SPSB2, an E3 ubiquitin ligase. Nitric Oxide 2021; 113-114:1-6. [PMID: 33862200 DOI: 10.1016/j.niox.2021.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/12/2021] [Accepted: 04/09/2021] [Indexed: 11/28/2022]
Abstract
Relatively high concentration of nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS) in response to a variety of stimuli is a source of reactive nitrogen species, an important weapon of host innate immune defense. The SPRY domain-containing SOCS box protein 2 (SPSB2) is an E3 ubiquitin ligase that regulates the lifetime of iNOS. SPSB2 interacts with the N-terminal region of iNOS via a binding site on the SPRY domain of SPSB2, and recruits an E3 ubiquitin ligase complex to polyubiquitinate iNOS, leading to its proteasomal degradation. Although critical residues for the SPSB2-iNOS interaction have been identified, structural basis for the interaction remains to be explicitly determined. In this study, we have determined a crystal structure of the N-terminal region of iNOS in complex with the SPRY domain of SPSB2 at 1.24 Å resolution. We have resolved the roles of some flanking residues, whose contribution to the SPSB2-iNOS interaction was structurally unclear previously. Furthermore, we have evaluated the effects of SPSB2 inhibitors on NO production using transient transfection and cell-penetrating peptide approaches, and found that such inhibitors can elevate NO production in RAW264.7 macrophages. These results thus provide a useful basis for the development of potent SPSB2 inhibitors as well as recruiting ligands for proteolysis targeting chimera (PROTAC) design.
Collapse
Affiliation(s)
- Kefa Li
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Tingting You
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Panqi Zhao
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Yanhong Luo
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Danting Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Huan Wei
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Yuhui Wang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Jinjin Yang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Xueyan Guan
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Zhihe Kuang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, 510632, China; Guangdong Provincial Biotechnology Drug and Engineering Technology Research Center, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China.
| |
Collapse
|
102
|
New insights on nitric oxide: Focus on animal models of schizophrenia. Behav Brain Res 2021; 409:113304. [PMID: 33865887 DOI: 10.1016/j.bbr.2021.113304] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a devastating complex disorder characterised by a constellation of behavioral deficits with the underlying mechanisms not fully known. Nitric oxide (NO) has emerged as a key signaling molecule implicated in schizophrenia. Three nitric oxide sinthases (NOS), endothelial, neuronal, and inducible, release NO within the cell. Animal models of schizophrenia are grouped in four groups, neurovedelopmental, glutamatergic, dopaminergic and genetic. In this review, we aim to evaluate changes in NO levels in animal models of schizophrenia and the resulting long-lasting behavioral and neural consequences. In particular, NO levels are substantially modified, region-specific, in various neurodevelopmental models, e.g. bilateral excitotoxic lesion of the ventral hippocampus (nVHL), maternal immune activation and direct NO manipulations early in development, among others. In regards to glutamatergic models of schizophrenia, phencyclidine (PCP) administration increases NO levels in the prefrontal cortex (PFC) and ventral hippocampus. As far as genetic models are concerned, neuronal NOS knock-out mice display schizophrenia-related behaviors. Administration of NO donors can reverse schizophrenia-related behavioral deficits. While most modifications in NO are derived from neuronal NOS, recent evidence indicates that PCP treatment increases NO from the inducible NOS isoform. From a pharmacological perspective, treatment with various antipsychotics including clozapine, haloperidol and risperidone normalize NO levels in the PFC as well as improve behavioral deficits in nVHL rats. NO induced from the neuronal and inducible NOS is relevant to schizophrenia and warrants further research.
Collapse
|
103
|
Choi YK, Kim YM. Regulation of Endothelial and Vascular Functions by Carbon Monoxide via Crosstalk With Nitric Oxide. Front Cardiovasc Med 2021; 8:649630. [PMID: 33912601 PMCID: PMC8071856 DOI: 10.3389/fcvm.2021.649630] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Carbon monoxide (CO), generated by heme oxygenase (HO), has been considered a signaling molecule in both the cardiovascular and central nervous systems. The biological function of the HO/CO axis is mostly related to other gaseous molecules, including nitric oxide (NO), which is synthesized by nitric oxide synthase (NOS). Healthy blood vessels are essential for the maintenance of tissue homeostasis and whole-body metabolism; however, decreased or impaired vascular function is a high-risk factor of cardiovascular and neuronal diseases. Accumulating evidence supports that the interplay between CO and NO plays a crucial role in vascular homeostasis and regeneration by improving endothelial function. Moreover, endothelial cells communicate with neighboring cells, such as, smooth muscle cells, immune cells, pericytes, and astrocytes in the periphery and neuronal vascular systems. Endogenous CO could mediate the cell-cell communication and improve the physiological functions of the cardiovascular and neurovascular systems via crosstalk with NO. Thus, a forward, positive feedback circuit between HO/CO and NOS/NO pathways can maintain cardiovascular and neurovascular homeostasis and prevent various human diseases. We discussed the crucial role of CO-NO crosstalk in the cardiovascular and neurovascular systems.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
104
|
Sun C, Zhang Y, Liu L, Liu X, Li B, Jin C, Lin X. Molecular functions of nitric oxide and its potential applications in horticultural crops. HORTICULTURE RESEARCH 2021; 8:71. [PMID: 33790257 PMCID: PMC8012625 DOI: 10.1038/s41438-021-00500-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 05/04/2023]
Abstract
Nitric oxide (NO) regulates plant growth, enhances nutrient uptake, and activates disease and stress tolerance mechanisms in most plants, making NO a potential tool for use in improving the yield and quality of horticultural crop species. Although the use of NO in horticulture is still in its infancy, research on NO in model plant species has provided an abundance of valuable information on horticultural crop species. Emerging evidence implies that the bioactivity of NO can occur through many potential mechanisms but occurs mainly through S-nitrosation, the covalent and reversible attachment of NO to cysteine thiol. In this context, NO signaling specifically affects crop development, immunity, and environmental interactions. Moreover, NO can act as a fumigant against a wide range of postharvest diseases and pests. However, for effective use of NO in horticulture, both understanding and exploring the biological significance and potential mechanisms of NO in horticultural crop species are critical. This review provides a picture of our current understanding of how NO is synthesized and transduced in plants, and particular attention is given to the significance of NO in breaking seed dormancy, balancing root growth and development, enhancing nutrient acquisition, mediating stress responses, and guaranteeing food safety for horticultural production.
Collapse
Affiliation(s)
- Chengliang Sun
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental & Resource Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Yuxue Zhang
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental & Resource Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Lijuan Liu
- Interdisciplinary Research Academy, Zhejiang Shuren University, 310015, Hangzhou, China
| | - Xiaoxia Liu
- Zhejiang Provincial Cultivated Land Quality and Fertilizer Administration Station, Hangzhou, China
| | - Baohai Li
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental & Resource Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Chongwei Jin
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental & Resource Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Xianyong Lin
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental & Resource Sciences, Zhejiang University, 310058, Hangzhou, China.
| |
Collapse
|
105
|
van Nispen J, Voigt M, Song E, Armstrong A, Fedorova M, Murali V, Krebs J, Samaddar A, Manithody C, Jain A. Parenteral Nutrition and Cardiotoxicity. Cardiovasc Toxicol 2021; 21:265-271. [PMID: 33554318 DOI: 10.1007/s12012-021-09638-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/27/2021] [Indexed: 12/30/2022]
Abstract
Parenteral nutrition (PN) is a life-saving nutritional therapy for those situations when patients are unable to receive enteral nutrition. However, despite a multitude of benefits offered by PN, it is associated with a variety of side effects, most notably parenteral nutrition-associated liver disease (PNALD). Adverse effects of PN on other organ systems, such as brain and cardiovascular system, have been poorly studied. There have been several case reports, studies, and a recent animal study highlighting cardiotoxic effects of PN; however, much remains unclear about the underlying mechanisms causing cardiac damage. In this review, we propose a series of potential mechanisms behind PN-associated heart injury, and we provide an overview of therapeutic strategies and recent scientific advances.
Collapse
Affiliation(s)
- Johan van Nispen
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA.
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA.
| | - Marcus Voigt
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| | - Eric Song
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| | - Austin Armstrong
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| | - Margarita Fedorova
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| | - Vidul Murali
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| | - Joseph Krebs
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| | - Ashish Samaddar
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| | | | - Ajay Jain
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, MO, 63103, USA
| |
Collapse
|
106
|
Skeletal muscle redox signaling in rheumatoid arthritis. Clin Sci (Lond) 2021; 134:2835-2850. [PMID: 33146370 PMCID: PMC7642299 DOI: 10.1042/cs20190728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovitis and the presence of serum autoantibodies. In addition, skeletal muscle weakness is a common comorbidity that contributes to inability to work and reduced quality of life. Loss in muscle mass cannot alone account for the muscle weakness induced by RA, but instead intramuscular dysfunction appears as a critical factor underlying the decreased force generating capacity for patients afflicted by arthritis. Oxidative stress and associated oxidative post-translational modifications have been shown to contribute to RA-induced muscle weakness in animal models of arthritis and patients with RA. However, it is still unclear how and which sources of reactive oxygen and nitrogen species (ROS/RNS) that are involved in the oxidative stress that drives the progression toward decreased muscle function in RA. Nevertheless, mitochondria, NADPH oxidases (NOX), nitric oxide synthases (NOS) and phospholipases (PLA) have all been associated with increased ROS/RNS production in RA-induced muscle weakness. In this review, we aim to cover potential ROS sources and underlying mechanisms of oxidative stress and loss of force production in RA. We also addressed the use of antioxidants and exercise as potential tools to counteract oxidative stress and skeletal muscle weakness.
Collapse
|
107
|
Virtuoso A, Giovannoni R, De Luca C, Gargano F, Cerasuolo M, Maggio N, Lavitrano M, Papa M. The Glioblastoma Microenvironment: Morphology, Metabolism, and Molecular Signature of Glial Dynamics to Discover Metabolic Rewiring Sequence. Int J Mol Sci 2021; 22:3301. [PMID: 33804873 PMCID: PMC8036663 DOI: 10.3390/ijms22073301] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Different functional states determine glioblastoma (GBM) heterogeneity. Brain cancer cells coexist with the glial cells in a functional syncytium based on a continuous metabolic rewiring. However, standard glioma therapies do not account for the effects of the glial cells within the tumor microenvironment. This may be a possible reason for the lack of improvements in patients with high-grade gliomas therapies. Cell metabolism and bioenergetic fitness depend on the availability of nutrients and interactions in the microenvironment. It is strictly related to the cell location in the tumor mass, proximity to blood vessels, biochemical gradients, and tumor evolution, underlying the influence of the context and the timeline in anti-tumor therapeutic approaches. Besides the cancer metabolic strategies, here we review the modifications found in the GBM-associated glia, focusing on morphological, molecular, and metabolic features. We propose to analyze the GBM metabolic rewiring processes from a systems biology perspective. We aim at defining the crosstalk between GBM and the glial cells as modules. The complex networking may be expressed by metabolic modules corresponding to the GBM growth and spreading phases. Variation in the oxidative phosphorylation (OXPHOS) rate and regulation appears to be the most important part of the metabolic and functional heterogeneity, correlating with glycolysis and response to hypoxia. Integrated metabolic modules along with molecular and morphological features could allow the identification of key factors for controlling the GBM-stroma metabolism in multi-targeted, time-dependent therapies.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | | | - Ciro De Luca
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Francesca Gargano
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Michele Cerasuolo
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
| | - Nicola Maggio
- Department of Neurology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel;
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5211401, Israel
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Michele Papa
- Laboratory of Neuronal Networks, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (F.G.); (M.C.); (M.P.)
- SYSBIO Centre of Systems Biology ISBE-IT, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
108
|
Szychowski KA, Skóra B, Wójtowicz AK. Triclosan affects the expression of nitric oxide synthases (NOSs), peroxisome proliferator-activated receptor gamma (PPARγ), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in mouse neocortical neurons in vitro. Toxicol In Vitro 2021; 73:105143. [PMID: 33722737 DOI: 10.1016/j.tiv.2021.105143] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/13/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022]
Abstract
Triclosan (TCS) is a well-known compound that can be found in disinfectants, personal care products. There is one publication concerning the involvement of PPARγ in the mechanism of action of TCS. It is known that activation of PPARγ regulates the expression of the NF-κB mediated inflammation by acting on nitric oxide synthase (NOS) genes. However, there are no studies demonstrating a relationship between the effects of TCS on the PPARγ signaling pathway, changes in NF-κB expression, and NOS isoform synthesis. Therefore, the aim of this study was to evaluate the effect of TCS on the expression of PPARγ, NF-κB, nNOS, iNOS, and eNOS in mouse neocortical neurons. In addition, the effects of co-administration of synthetic alpha-naphthoflavone (αNF) or beta-naphthoflavone (βNF) flavonoids and triclosan were investigated. Our results show that TCS alters PPARγ, NF-κB, iNOS, and eNOS expression in mouse neurons in vitro. After 48 h of exposure, TCS increased PPARγ expression and decreased NF-κB expression. Moreover, under the influence of TCS, the expression of iNOS was increased and at the same time the expression of nNOS was decreased, which was probably caused by high levels of ROS. The experiments have shown that both αNF and βNF are able to modulate the effects of TCS in primary cultures of mouse cortical neurons.
Collapse
Affiliation(s)
- Konrad A Szychowski
- Department of Biotechnology and Cell Biology, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland.
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Anna K Wójtowicz
- Department of Nutrition, Animal Biotechnology and Fisheries, Faculty of Animal Sciences, University of Agriculture, Adama Mickiewicza 24/28, 30-059 Kraków, Poland
| |
Collapse
|
109
|
Galan-Vasquez E, Perez-Rueda E. A landscape for drug-target interactions based on network analysis. PLoS One 2021; 16:e0247018. [PMID: 33730052 PMCID: PMC7968663 DOI: 10.1371/journal.pone.0247018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/30/2021] [Indexed: 12/30/2022] Open
Abstract
In this work, we performed an analysis of the networks of interactions between drugs and their targets to assess how connected the compounds are. For our purpose, the interactions were downloaded from the DrugBank database, and we considered all drugs approved by the FDA. Based on topological analysis of this interaction network, we obtained information on degree, clustering coefficient, connected components, and centrality of these interactions. We identified that this drug-target interaction network cannot be divided into two disjoint and independent sets, i.e., it is not bipartite. In addition, the connectivity or associations between every pair of nodes identified that the drug-target network is constituted of 165 connected components, where one giant component contains 4376 interactions that represent 89.99% of all the elements. In this regard, the histamine H1 receptor, which belongs to the family of rhodopsin-like G-protein-coupled receptors and is activated by the biogenic amine histamine, was found to be the most important node in the centrality of input-degrees. In the case of centrality of output-degrees, fostamatinib was found to be the most important node, as this drug interacts with 300 different targets, including arachidonate 5-lipoxygenase or ALOX5, expressed on cells primarily involved in regulation of immune responses. The top 10 hubs interacted with 33% of the target genes. Fostamatinib stands out because it is used for the treatment of chronic immune thrombocytopenia in adults. Finally, 187 highly connected sets of nodes, structured in communities, were also identified. Indeed, the largest communities have more than 400 elements and are related to metabolic diseases, psychiatric disorders and cancer. Our results demonstrate the possibilities to explore these compounds and their targets to improve drug repositioning and contend against emergent diseases.
Collapse
Affiliation(s)
- Edgardo Galan-Vasquez
- Departamento de Ingeniería de Sistemas Computacionales y Automatización, Instituto de Investigación en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Ciudad Universitaria, México City, México
| | - Ernesto Perez-Rueda
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Unidad Académica Yucatán, Mérida, Yucatán, México
| |
Collapse
|
110
|
Estes LM, Singha P, Singh S, Sakthivel TS, Garren M, Devine R, Brisbois EJ, Seal S, Handa H. Characterization of a nitric oxide (NO) donor molecule and cerium oxide nanoparticle (CNP) interactions and their synergistic antimicrobial potential for biomedical applications. J Colloid Interface Sci 2021; 586:163-177. [DOI: 10.1016/j.jcis.2020.10.081] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
|
111
|
Dao VTV, Elbatreek MH, Fuchß T, Grädler U, Schmidt HHHW, Shah AM, Wallace A, Knowles R. Nitric Oxide Synthase Inhibitors into the Clinic at Last. Handb Exp Pharmacol 2021; 264:169-204. [PMID: 32797331 DOI: 10.1007/164_2020_382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 1998 Nobel Prize in Medicine and Physiology for the discovery of nitric oxide, a nitrogen containing reactive oxygen species (also termed reactive nitrogen or reactive nitrogen/oxygen species) stirred great hopes. Clinical applications, however, have so far pertained exclusively to the downstream signaling of cGMP enhancing drugs such as phosphodiesterase inhibitors and soluble guanylate cyclase stimulators. All clinical attempts, so far, to inhibit NOS have failed even though preclinical models were strikingly positive and clinical biomarkers correlated perfectly. This rather casts doubt on our current way of target identification in drug discovery in general and our way of patient stratification based on correlating but not causal biomarkers or symptoms. The opposite, NO donors, nitrite and enhancing NO synthesis by eNOS/NOS3 recoupling in situations of NO deficiency, are rapidly declining in clinical relevance or hold promise but need yet to enter formal therapeutic guidelines, respectively. Nevertheless, NOS inhibition in situations of NO overproduction often jointly with enhanced superoxide (or hydrogen peroxide production) still holds promise, but most likely only in acute conditions such as neurotrauma (Stover et al., J Neurotrauma 31(19):1599-1606, 2014) and stroke (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019). Conversely, in chronic conditions, long-term inhibition of NOS might be too risky because of off-target effects on eNOS/NOS3 in particular for patients with cardiovascular risks or metabolic and renal diseases. Nitric oxide synthases (NOS) and their role in health (green) and disease (red). Only neuronal/type 1 NOS (NOS1) has a high degree of clinical validation and is in late stage development for traumatic brain injury, followed by a phase II safety/efficacy trial in ischemic stroke. The pathophysiology of NOS1 (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016) is likely to be related to parallel superoxide or hydrogen peroxide formation (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 114(46):12315-12320, 2017; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019) leading to peroxynitrite and protein nitration, etc. Endothelial/type 3 NOS (NOS3) is considered protective only and its inhibition should be avoided. The preclinical evidence for a role of high-output inducible/type 2 NOS (NOS2) isoform in sepsis, asthma, rheumatic arthritis, etc. was high, but all clinical development trials in these indications were neutral despite target engagement being validated. This casts doubt on the role of NOS2 in humans in health and disease (hence the neutral, black coloring).
Collapse
Affiliation(s)
- Vu Thao-Vi Dao
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Thomas Fuchß
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Ulrich Grädler
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Alan Wallace
- Health and Life Sciences, Coventry University, Coventry, UK
| | - Richard Knowles
- Knowles Consulting Ltd., The Stevenage Bioscience Catalyst, Stevenage, UK.
| |
Collapse
|
112
|
Nogales C, Grønning AGB, Sadegh S, Baumbach J, Schmidt HHHW. Network Medicine-Based Unbiased Disease Modules for Drug and Diagnostic Target Identification in ROSopathies. Handb Exp Pharmacol 2021; 264:49-68. [PMID: 32780286 DOI: 10.1007/164_2020_386] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Most diseases are defined by a symptom, not a mechanism. Consequently, therapies remain symptomatic. In reverse, many potential disease mechanisms remain in arbitrary search for clinical relevance. Reactive oxygen species (ROS) are such an example. It is an attractive hypothesis that dysregulation of ROS can become a disease trigger. Indeed, elevated ROS levels of various biomarkers have been correlated with almost every disease, yet after decades of research without any therapeutic application. We here present a first systematic, non-hypothesis-based approach to transform this field as a proof of concept for biomedical research in general. We selected as seed proteins 9 families with 42 members of clinically researched ROS-generating enzymes, ROS-metabolizing enzymes or ROS targets. Applying an unbiased network medicine approach, their first neighbours were connected, and, based on a stringent subnet participation degree (SPD) of 0.4, hub nodes excluded. This resulted in 12 distinct human interactome-based ROS signalling modules, while 8 proteins remaining unconnected. This ROSome is in sharp contrast to commonly used highly curated and integrated KEGG, HMDB or WikiPathways. These latter serve more as mind maps of possible ROS signalling events but may lack important interactions and often do not take different cellular and subcellular localization into account. Moreover, novel non-ROS-related proteins were part of these forming functional hybrids, such as the NOX5/sGC, NOX1,2/NOS2, NRF2/ENC-1 and MPO/SP-A modules. Thus, ROS sources are not interchangeable but associated with distinct disease processes or not at all. Module members represent leads for precision diagnostics to stratify patients with specific ROSopathies for precision intervention. The upper panel shows the classical approach to generate hypotheses for a role of ROS in a given disease by focusing on ROS levels and to some degree the ROS type or metabolite. Low levels are considered physiological; higher amounts are thought to cause a redox imbalance, oxidative stress and eventually disease. The source of ROS is less relevant; there is also ROS-induced ROS formation, i.e. by secondary sources (see upwards arrow). The non-hypothesis-based network medicine approach uses genetically or otherwise validated risk genes to construct disease-relevant signalling modules, which will contain also ROS targets. Not all ROS sources will be relevant for a given disease; some may not be disease relevant at all. The three examples show (from left to right) the disease-relevant appearance of an unphysiological ROS modifier/toxifier protein, ROS target or ROS source.
Collapse
Affiliation(s)
- Cristian Nogales
- Department of Pharmacology and Personalised Medicine, Maastricht University, Maastricht, The Netherlands.
| | - Alexander G B Grønning
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Sepideh Sadegh
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Jan Baumbach
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark.,Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
113
|
Akinrinde AS, Hameed HO. Glycine and L-Arginine supplementation ameliorates gastro-duodenal toxicity in a rat model of NSAID (Diclofenac)-gastroenteropathy via inhibition of oxidative stress. J Basic Clin Physiol Pharmacol 2021; 33:285-295. [PMID: 33559459 DOI: 10.1515/jbcpp-2020-0307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/22/2020] [Indexed: 01/09/2023]
Abstract
OBJECTIVES This study examined the possible protective roles of exogenous glycine (Gly) and L-Arginine (l-Arg) against Diclofenac (DIC)-induced gastro-duodenal damage in rats. METHODS Rats were divided into Group A (control), Group B (DIC group) and Groups C-F which were pre-treated for five days with Gly1 (250 mg/kg), Gly2 (500 mg/kg), l-Arg1 (200 mg/kg) and l-Arg2 (400 mg/kg), respectively, before co-treatment with DIC for another three days. Hematological, biochemical and histopathological analyses were then carried out. RESULTS DIC produced significant (p<0.05) reduction in PCV (13.82%), Hb (46.58%), RBC (30.53%), serum total protein (32.72%), albumin (28.44%) and globulin (38.01%) along with significant (p<0.05) elevation of serum MPO activity (83.30%), when compared with control. In addition, DIC increased gastric H2O2 and MDA levels by 33.93 and 48.59%, respectively, while the duodenal levels of the same parameters increased by 19.43 and 85.56%, respectively. Moreover, SOD, GPx and GST activities in the DIC group were significantly (p<0.05) reduced in the stomach (21.12, 24.35 and 51.28%, respectively) and duodenum (30.59, 16.35 and 37.90%, respectively), compared to control. Treatment with Gly and l-Arg resulted in significant amelioration of the DIC-induced alterations although l-Arg produced better amelioration of RBC (29.78%), total protein (10.12%), albumin (9.93%) and MPO (65.01%), compared to the DIC group. The protective effects of both amino acids against oxidative stress parameters and histological lesions were largely similar. CONCLUSIONS The data from this study suggest that Gly or l-Arg prevented DIC-induced gastro-duodenal toxicity and might, therefore be useful in improving the therapeutic index of DIC.
Collapse
Affiliation(s)
- Akinleye Stephen Akinrinde
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Halimot Olawalarami Hameed
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
114
|
Tan Y, Li Q, Zhao Y, Wei H, Wang J, Baker CJ, Liu Q, Wei W. Integration of metabolomics and existing omics data reveals new insights into phytoplasma-induced metabolic reprogramming in host plants. PLoS One 2021; 16:e0246203. [PMID: 33539421 PMCID: PMC7861385 DOI: 10.1371/journal.pone.0246203] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/14/2021] [Indexed: 12/03/2022] Open
Abstract
Phytoplasmas are cell wall-less bacteria that induce abnormal plant growth and various diseases, causing severe economic loss. Phytoplasmas are highly dependent on nutrients imported from host cells because they have lost many genes involved in essential metabolic pathways during reductive evolution. However, metabolic crosstalk between phytoplasmas and host plants and the mechanisms of phytoplasma nutrient acquisition remain poorly understood. In this study, using metabolomics approach, sweet cherry virescence (SCV) phytoplasma-induced metabolite alterations in sweet cherry trees were investigated. A total of 676 metabolites were identified in SCV phytoplasma-infected and mock inoculated leaves, of which 187 metabolites were differentially expressed, with an overwhelming majority belonging to carbohydrates, fatty acids/lipids, amino acids, and flavonoids. Available omics data of interactions between plant and phytoplasma were also deciphered and integrated into the present study. The results demonstrated that phytoplasma infection promoted glycolysis and pentose phosphate pathway activities, which provide energy and nutrients, and facilitate biosynthesis of necessary low-molecular metabolites. Our findings indicated that phytoplasma can induce reprograming of plant metabolism to obtain nutrients for its own replication and infection. The findings from this study provide new insight into interactions of host plants and phytoplasmas from a nutrient acquisition perspective.
Collapse
Affiliation(s)
- Yue Tan
- Shandong Institute of Pomology, Taian, China
| | - Qingliang Li
- College of Life Sciences, Zaozhuang University, Zaozhuang, China
| | - Yan Zhao
- United States Department of Agriculture, Molecular Plant Pathology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, Beltsville, MD, United States of America
| | - Hairong Wei
- Shandong Institute of Pomology, Taian, China
| | - Jiawei Wang
- Shandong Institute of Pomology, Taian, China
| | - Con Jacyn Baker
- United States Department of Agriculture, Molecular Plant Pathology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, Beltsville, MD, United States of America
| | | | - Wei Wei
- United States Department of Agriculture, Molecular Plant Pathology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, Beltsville, MD, United States of America
| |
Collapse
|
115
|
Souza JFD, Mello ADA, Portal TM, Nunes-da-Fonseca R, Monteiro de Barros C. Novel insights about the ascidian dopamine system: Pharmacology and phylogenetics of catecholaminergic receptors on the Phallusia nigra immune cells. FISH & SHELLFISH IMMUNOLOGY 2021; 109:41-50. [PMID: 33285170 DOI: 10.1016/j.fsi.2020.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/22/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
Dopamine (DA) is an important molecule that plays a role in the nervous and immune systems. DA is produced by a wide variety of animals and it is considered one of the oldest neurotransmitters. However, its specific function in immune cells has not been completely revealed. In a group of chordate animals, the ascidians, DA is reported to be produced by cells in the central nervous system (CNS); however, no dopaminergic receptor in their genomes has been described until now. Because this is an integrating characteristic of the ascidian dopamine system, here it was investigated the pharmacology, function, and phylogeny of DA and dopaminergic receptors (DRs) in the modulation of nitric oxide (NO) in the Phallusia nigra immune cells. The data disclosed, for the first time, that DA modulates NO production by immune cells. Its modulation probably occurs though adrenergic receptors, which display a special characteristic, in that they are capable of binding to noradrenaline (NA) and DA. A pharmacological analysis revealed that receptors present on the ascidian immune cells showed a high affinity to butaclamol, a non-selective D2-class receptor, increasing NO production. In addition, calcium intracellular mobilization was observed when DA was added to immune cells. In conclusion, the data revealed novel insights about the presence of catecholaminergic receptors (CRs) on the P. nigra immune cells, indicating that ascidian CRs have special pharmacological characteristics that are worth highlighting from an evolutionary point of view.
Collapse
Affiliation(s)
- Jessica Fernandes de Souza
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ, Brazil; Pós-Graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Campus UFRJ- Macaé, Macaé, RJ, Brazil
| | - Andressa de Abreu Mello
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ, Brazil; Pós-Graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Campus UFRJ- Macaé, Macaé, RJ, Brazil; Pós-Graduação em Ciências Biológicas, Biofísica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro- UFRJ, Rio de Janeiro, RJ, Brazil
| | - Taynan Motta Portal
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ, Brazil; Pós-Graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Campus UFRJ- Macaé, Macaé, RJ, Brazil
| | - Rodrigo Nunes-da-Fonseca
- Laboratório Integrado de Ciências Morfofuncionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ, Brazil
| | - Cintia Monteiro de Barros
- Laboratório Integrado de Biociências Translacionais, Instituto de Biodiversidade e Sustentabilidade - NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ, Brazil; Pós-Graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Campus UFRJ- Macaé, Macaé, RJ, Brazil.
| |
Collapse
|
116
|
Girotti AW. Nitric Oxide-elicited Resistance to Antitumor Photodynamic Therapy via Inhibition of Membrane Free Radical-mediated Lipid Peroxidation. Photochem Photobiol 2021; 97:653-663. [PMID: 33369741 DOI: 10.1111/php.13373] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022]
Abstract
This review focuses on the ability of nitric oxide (NO) to antagonize antitumor photodynamic therapy (PDT). NO's anti-PDT effects were recognized relatively recently and require a better mechanistic understanding for developing new strategies to improve PDT efficacy. Many PDT sensitizers (PSs) are amphiphilic and tend to localize in membrane compartments of tumor cells. Unsaturated lipids in these compartments can undergo peroxidative degradation after PS photoactivation. Primary Type I (free radical) vs. Type II (singlet oxygen) photochemistry of lipid peroxidation is discussed, along with light-independent turnover of primary lipid hydroperoxides to free radical species. Chain lipid peroxidation mediated by the latter exacerbates membrane damage and cytotoxicity after a PDT challenge. Our studies have shown that NO from chemical donors can suppress chain peroxidation by intercepting lipid-derived free radical intermediates, thereby protecting cancer cells against photokilling. More recent evidence has revealed that inducible NO synthase (iNOS) is dramatically upregulated in several cancer cell types after a photodynamic challenge, and that iNOS-derived NO enhances resistance as well as growth and migratory aggressiveness of surviving cells. Chain breaking by NO and other possible NO-based resistance mechanisms are discussed, along with novel pharmacologic approaches for overcoming these negative effects.
Collapse
Affiliation(s)
- Albert W Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
117
|
Paulo M, Costa DEFR, Bonaventura D, Lunardi CN, Bendhack LM. Nitric Oxide Donors as Potential Drugs for the Treatment of Vascular Diseases Due to Endothelium Dysfunction. Curr Pharm Des 2021; 26:3748-3759. [PMID: 32427079 DOI: 10.2174/1381612826666200519114442] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/07/2020] [Indexed: 11/22/2022]
Abstract
Endothelial dysfunction and consequent vasoconstriction are a common condition in patients with hypertension and other cardiovascular diseases. Endothelial cells produce and release vasodilator substances that play a pivotal role in normal vascular tone. The mechanisms underlying endothelial dysfunction are multifactorial. However, enhanced reactive oxygen species (ROS) production and consequent vasoconstriction instead of endothelium-derived relaxant generation and consequent vasodilatation contribute to this dysfunction considerably. The main targets of the drugs that are currently used to treat vascular diseases concerning enzyme activities and protein functions that are impaired by endothelial nitric oxide synthase (eNOS) uncoupling and ROS production. Nitric oxide (NO) bioavailability can decrease due to deficient NO production by eNOS and/or NO release to vascular smooth muscle cells, which impairs endothelial function. Considering the NO cellular mechanisms, tackling the issue of eNOS uncoupling could avoid endothelial dysfunction: provision of the enzyme cofactor tetrahydrobiopterin (BH4) should elicit NO release from NO donors, to activate soluble guanylyl cyclase. This should increase cyclic guanosine-monophosphate (cGMP) generation and inhibit phosphodiesterases (especially PDE5) that selectively degrade cGMP. Consequently, protein kinase-G should be activated, and K+ channels should be phosphorylated and activated, which is crucial for cell membrane hyperpolarization and vasodilation and/or inhibition of ROS production. The present review summarizes the current concepts about the vascular cellular mechanisms that underlie endothelial dysfunction and which could be the target of drugs for the treatment of patients with cardiovascular disease.
Collapse
Affiliation(s)
- Michele Paulo
- Department Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirao Preto- University of Sao Paulo Av. Do Cafe SN, Brazil
| | - Daniela E F R Costa
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Daniella Bonaventura
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Claure N Lunardi
- Laboratory of Photochemistry and Nanobiotechnology, University of Brasilia, Brasilia, Brazil
| | - Lusiane M Bendhack
- Department Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirao Preto- University of Sao Paulo Av. Do Cafe SN, Brazil
| |
Collapse
|
118
|
Large magnitude of force leads to NO-mediated cell shrinkage in single osteocytes implying an initial apoptotic response. J Biomech 2021; 117:110245. [PMID: 33493709 DOI: 10.1016/j.jbiomech.2021.110245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/06/2020] [Accepted: 01/03/2021] [Indexed: 11/22/2022]
Abstract
Damage accumulation in the bone under continuous daily loading causes local mechanical overloading known to induce osteocyte apoptosis, which promotes bone resorption to repair bone damage. However, only a few studies have investigated the mechanism of apoptosis in mechanically overloaded osteocytes. As mechanically stimulated osteocytes produce nitric oxide (NO), which triggers apoptosis in various cell types, we aimed to elucidate the mechanism underlying apoptosis in mechanically overloaded osteocytes, focusing on intracellular NO. To investigate the effects of force magnitude on apoptosis and intracellular NO production, we isolated osteocytes from DMP1-EGFP mice and subjected them to quantitative local forces via fibronectin-coated micro beads targeting integrin on the cell surface using a magnetic tweezer. Cell shrinkage was microscopically examined, and intracellular NO production was visualized using DAR-4 M. Mechanical stimulation revealed relationships between force magnitude, apoptosis, and intracellular NO production. The application of a smaller force resulted in no significant cell shrinkage or intracellular NO production; however, a larger force caused a rapid increase in intracellular NO production followed by cell shrinkage. Besides, intracellular NOS (NO synthase) inhibition and NO donation revealed the pro-apoptotic roles of NO in osteocytes. L-NAME (NOS inhibitor)-treated cells displayed no significant shrinkage under a larger force, whereas SNP (NO donor)-treated cells showed cell shrinkage and Annexin V fluorescence, indicating apoptosis. Collectively, our study demonstrates that larger force leads to NO production-mediated osteocyte shrinkage, implying an initial apoptotic response and highlighting the importance of NO production in bone damage.
Collapse
|
119
|
Vasorelaxant effect of water fraction of Labisia Pumila and its mechanisms in spontaneously hypertensive rats aortic ring preparation. INTERNATIONAL JOURNAL CARDIOLOGY HYPERTENSION 2021; 4:100024. [PMID: 33447753 PMCID: PMC7803023 DOI: 10.1016/j.ijchy.2020.100024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/16/2019] [Accepted: 12/25/2019] [Indexed: 11/24/2022]
Abstract
Introduction Labisia pumila has been reported to possess activities including antioxidant, anti-aging and anti-cancer but there is no report on its vasorelaxant effects. Objective This study aims to fractionate water extract of Labisia pumila, identify the compound(s) involved and elucidate the possible mechanism(s) of its vasorelaxant effects. Methods Water extract of Labisia pumila was subjected to liquid-liquid extraction to obtain ethyl acetate, n-butanol and water fractions. In SHR aortic ring preparations, water fraction (WF-LPWE) was established as the most potent fraction for vasorelaxation. The pharmacological mechanisms of the vasorelaxant effect of WF-LPWE were investigated with and without the presence of various inhibitors. The cumulative dose-response curves of potassium chloride (KCl)-induced contractions were conducted to study the possible mechanisms of WF-LPWE in reducing vasoconstriction. Results WF-LPWE produced dose-dependent vasorelaxant effect in endothelium-denuded aortic ring and showed non-competitive inhibition of dose-response curves of PE-induced contraction, and at its higher concentrations reduced KCl-induced contraction. 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) significantly inhibited vasorelaxant effect of WF-LPWE. WF-LPWE significantly reduced the release of intracellular calcium ion (Ca2+) from the intracellular stores and suppressed the calcium chloride (CaCal2)-induced contraction. Nω-nitro-L-arginine methyl ester (L-NAME), methylene blue, indomethacin and atropine did not influence the vasorelaxant effects of WF-LPWE. Conclusion WF-LPWE exerts its vasorelaxant effect independently of endothelium and possibly by inhibiting the release of calcium from intracellular calcium stores, receptor-operated calcium channels and formation of inositol 1,4,5- triphosphate. WF-LPWE vasorelaxant effect may also mediated via nitric oxide-independent direct involvement of soluble guanylate cyclase (sGC)/ cyclic guanosine monophosphate (cGMP) pathways.
Collapse
|
120
|
Fultang N, Li X, Li T, Chen YH. Myeloid-Derived Suppressor Cell Differentiation in Cancer: Transcriptional Regulators and Enhanceosome-Mediated Mechanisms. Front Immunol 2021; 11:619253. [PMID: 33519825 PMCID: PMC7840597 DOI: 10.3389/fimmu.2020.619253] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 01/16/2023] Open
Abstract
Myeloid-derived Suppressor Cells (MDSCs) are a sub-population of leukocytes that are important for carcinogenesis and cancer immunotherapy. During carcinogenesis or severe infections, inflammatory mediators induce MDSCs via aberrant differentiation of myeloid precursors. Although several transcription factors, including C/EBPβ, STAT3, c-Rel, STAT5, and IRF8, have been reported to regulate MDSC differentiation, none of them are specifically expressed in MDSCs. How these lineage-non-specific transcription factors specify MDSC differentiation in a lineage-specific manner is unclear. The recent discovery of the c-Rel-C/EBPβ enhanceosome in MDSCs may help explain these context-dependent roles. In this review, we examine several transcriptional regulators of MDSC differentiation, and discuss the concept of non-modular regulation of MDSC signature gene expression by transcription factors such as c-Rel and C/EBPß.
Collapse
Affiliation(s)
- Norman Fultang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | | | - Youhai H. Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
121
|
Vedenko A, Panara K, Goldstein G, Ramasamy R, Arora H. Tumor Microenvironment and Nitric Oxide: Concepts and Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1277:143-158. [PMID: 33119871 DOI: 10.1007/978-3-030-50224-9_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cancer tissue exists not as a single entity, but as a combination of different cellular phenotypes which, taken together, dramatically contribute to the entirety of their ecosystem, collectively termed as the tumor microenvironment (TME). The TME is composed of both immune and nonimmune cell types, stromal components, and vasculature-all of which cooperate to promote cancer progression. Not all immune cells, however, are immune-suppressive; some of them can promote the immune microenvironment to fight the invading and uncontrollably dividing cell populations at the initial stages of tumor growth. Yet, many of these processes and cellular phenotypes fall short, and the immune ecosystem more often than not ends up stabilizing in favor of the "resistant" resident cells that begin clonal expansion and may progress to metastatic forms. Stromal components, making up the extracellular matrix and basement membrane, are also not the most innocuous: CAFs embedded throughout secrete proteases that allow the onset of one of the most invasive processes-angiogenesis-through destruction of the ECM and the basement membrane. Vasculature formation, because of angiogenesis, is the largest invader of the TME and the reason metastasis happens. Vasculature is so sporadic and omnipresent in the TME that most drug therapies are mainly focused on stopping this uncontrollable process. As the tumor continues to grow, different processes are constantly supplying it with the ingredients favorable for tumor progression and eventual metastasis. For example, angiogenesis promotes blood vessel formation that will allow the bona fide escape of tumor cells to take place. Another process like hypoxia will present itself in several forms throughout the tumor (mild or acute, cycling or permanent), starting mechanisms such as epithelial to mesenchymal transitions (EMT) of resident cells and inadvertently placing the cells in such a stressful condition that production of ROS and DNA damage is unavoidable. DNA damage can induce mutagenicity while allowing resistant cells to survive. This is where drugs and treatments can subsequently suffer in effectiveness. Finally, another molecule has just surfaced as being a very important player in the TME: nitric oxide. Often overlooked and equated with ROS and initially assigned in the category of pathogenic molecules, nitric oxide can definitely do some damage by causing metabolic reprogramming and promotion of immunosuppressive phenotypes at low concentrations. However, its actions seem to be extremely dose-dependent, and this issue has become a hot target of current treatment goals. Shockingly, nitric oxide, although omnipresent in the TME, can have a positive effect on targeting the TME broadly. Thus, while the TME is a myriad of cellular phenotypes and a combination of different tumor-promoting processes, each process is interconnected into one whole: the tumor microenvironment.
Collapse
Affiliation(s)
- Anastasia Vedenko
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Kush Panara
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Gabriella Goldstein
- College of Health Professions and Sciences, University of Central Florida, Orlando, FL, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Himanshu Arora
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
122
|
Alsharabasy AM, Pandit A, Farràs P. Recent Advances in the Design and Sensing Applications of Hemin/Coordination Polymer-Based Nanocomposites. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2003883. [PMID: 33217074 DOI: 10.1002/adma.202003883] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/10/2020] [Indexed: 06/11/2023]
Abstract
The fabrication of biomimetic catalysts as substituents for enzymes is of critical interest in the field due to the problems associated with the extraction, purification, and storage of enzymes in sensing applications. Of these mimetics, hemin/coordination polymer-based nanocomposites, mainly hemin/metal-organic frameworks (MOF), have been developed for various biosensing applications because of the unique properties of each component, while trying to mimic the normal biological functions of heme within the protein milieu of enzymes. This critical review first discusses the different catalytic functions of heme in the body in the form of enzyme/protein structures. The properties of hemin dimerization are then elucidated with the supposed models of hemin oxidation. After that, the progress in the fabrication of hemin/MOF nanocomposites for the sensing of diverse biological molecules is discussed. Finally, the challenges in developing this type of composites are examined as well as possible proposals for future directions to enhance the sensing performance in this field further.
Collapse
Affiliation(s)
- Amir M Alsharabasy
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, H91W2TY, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, H91W2TY, Ireland
| | - Pau Farràs
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, H91W2TY, Ireland
- School of Chemistry, Ryan Institute, National University of Ireland Galway, Galway, H91CF50, Ireland
| |
Collapse
|
123
|
Zhou W, Zhou R, Li N, Chen Y, Pei Y, Han L, Ren J. Vasorelaxation effect of oxysophoridine on isolated thoracicc aorta rings of rats. CHINESE J PHYSIOL 2021; 64:274-280. [DOI: 10.4103/cjp.cjp_60_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
124
|
Mir JM, Maurya RC. Nitric oxide as a therapeutic option for COVID-19 treatment: a concise perspective. NEW J CHEM 2021. [DOI: 10.1039/d0nj03823g] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Among several possible therapies applicable for treating COVID-19, nitric oxide therapy has also gained considerable interest. This article describes the same with mechanistic details.
Collapse
Affiliation(s)
- Jan Mohammad Mir
- Coordination, Metallopharmaceutical and Computational Laboratory
- Department of PG Studies and Research in Chemistry and Pharmacy
- R. D. University
- Jabalpur
- India
| | - Ram Charitra Maurya
- Coordination, Metallopharmaceutical and Computational Laboratory
- Department of PG Studies and Research in Chemistry and Pharmacy
- R. D. University
- Jabalpur
- India
| |
Collapse
|
125
|
Girotti AW, Fahey JM, Korytowski W. Negative effects of tumor cell nitric oxide on anti-glioblastoma photodynamic therapy. JOURNAL OF CANCER METASTASIS AND TREATMENT 2020; 6:52. [PMID: 33564720 PMCID: PMC7869587 DOI: 10.20517/2394-4722.2020.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glioblastomas are highly aggressive brain tumors that can persist after exposure to conventional chemotherapy or radiotherapy. Nitric oxide (NO) produced by inducible NO synthase (iNOS/NOS2) in these tumors is known to foster malignant cell proliferation, migration, and invasion as well as resistance to chemo- and radiotherapy. Minimally invasive photodynamic therapy (PDT) sensitized by 5-aminolevulinic acid (ALA)-induced protoporphyrin IX (PpIX) is a highly effective anti-glioblastoma modality, but it is also subject to NO-mediated resistance. Studies by the authors have revealed that glioblastoma U87 and U251 cells use endogenous iNOS/NO to not only resist photokilling after an ALA/light challenge, but also to promote proliferation and migration/invasion of surviving cells. Stress-upregulated iNOS/NO was found to play a major role in these negative responses to PDT-like treatment. Our studies have revealed a tight network of upstream signaling events leading to iNOS induction in photostressed cells and transition to a more aggressive phenotype. These events include activation or upregulation of pro-survival/ pro-expansion effector proteins such as NF-κB, phosphoinositide-3-kinase (PI3K), protein kinase-B (Akt), p300, Survivin, and Brd4. In addition to this upstream signaling and its regulation, pharmacologic approaches for directly suppressing iNOS at its activity vs. transcriptional level are discussed. One highly effective agent in the latter category is bromodomain and extra-terminal (BET) inhibitor, JQ1, which was found to minimize iNOS upregulation in photostressed U87 cells. By acting similarly at the clinical level, a BET inhibitor such as JQ1 should markedly improve the efficacy of anti-glioblastoma PDT.
Collapse
Affiliation(s)
- Albert W. Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Jonathan M. Fahey
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Witold Korytowski
- Department of Biophysics, Jagiellonian University, Krakow 30-387, Poland
| |
Collapse
|
126
|
Wigner P, Synowiec E, Jóźwiak P, Czarny P, Bijak M, Białek K, Szemraj J, Gruca P, Papp M, Śliwiński T. The Effect of Chronic Mild Stress and Escitalopram on the Expression and Methylation Levels of Genes Involved in the Oxidative and Nitrosative Stresses as Well as Tryptophan Catabolites Pathway in the Blood and Brain Structures. Int J Mol Sci 2020; 22:ijms22010010. [PMID: 33374959 PMCID: PMC7792593 DOI: 10.3390/ijms22010010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Previous studies suggest that depression may be associated with reactive oxygen species overproduction and disorders of the tryptophan catabolites pathway. Moreover, one-third of patients do not respond to conventional pharmacotherapy. Therefore, the study investigates the molecular effect of escitalopram on the expression of Cat, Gpx1/4, Nos1/2, Tph1/2, Ido1, Kmo, and Kynu and promoter methylation in the hippocampus, amygdala, cerebral cortex, and blood of rats exposed to CMS (chronic mild stress). The animals were exposed to CMS for two or seven weeks followed by escitalopram treatment for five weeks. The mRNA and protein expression of the genes were analysed using the TaqMan Gene Expression Assay and Western blotting, while the methylation was determined using methylation-sensitive high-resolution melting. The CMS caused an increase of Gpx1 and Nos1 mRNA expression in the hippocampus, which was normalised by escitalopram administration. Moreover, Tph1 and Tph2 mRNA expression in the cerebral cortex was increased in stressed rats after escitalopram therapy. The methylation status of the Cat promoter was decreased in the hippocampus and cerebral cortex of the rats after escitalopram therapy. The Gpx4 protein levels were decreased following escitalopram compared to the stressed/saline group. It appears that CMS and escitalopram influence the expression and methylation of the studied genes.
Collapse
Affiliation(s)
- Paulina Wigner
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
| | - Ewelina Synowiec
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
| | - Paweł Jóźwiak
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland;
| | - Piotr Czarny
- Department of Medical Biochemistry, Medical University of Lodz, 90-647 Lodz, Poland; (P.C.); (J.S.)
| | - Michał Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland;
| | - Katarzyna Białek
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 90-647 Lodz, Poland; (P.C.); (J.S.)
| | - Piotr Gruca
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland; (P.G.); (M.P.)
| | - Mariusz Papp
- Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland; (P.G.); (M.P.)
| | - Tomasz Śliwiński
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-136 Lodz, Poland; (P.W.); (E.S.); (K.B.)
- Correspondence: ; Tel.: +48-42-635-44-86; Fax: +48-42-635-44-84
| |
Collapse
|
127
|
The effects of regular aerobic exercise training on blood nitric oxide levels and oxidized LDL and the role of eNOS intron 4a/b polymorphism. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165913. [PMID: 32795498 DOI: 10.1016/j.bbadis.2020.165913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/03/2020] [Accepted: 07/31/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Nitric oxide (NO), oxidized LDL (OxLDL) and endothelial nitric oxide synthase intron 4a/b polymorphism (eNOSP) are related to atherosclerosis (AS). The present study investigated the effects of regular aerobic exercise training on the mentioned risk factors as well as blood lipids and lipoproteins (BLLPs) and the role of eNOSP, which is unclear. METHODS The study was participated by 46 well trained male soccer referees as the athletic group (AG, age; 23.26 ± 2.84 years) and 43 sedentary controls (CG, age; 23.16 ± 3.28 years). Yoyo intermittent endurance (Yoyo IE-2 test) was performed to measure aerobic endurance levels of the participants. Serum NO, eNOS and oxidized LDL (OxLDL) levels (by ELISA method) and total oxidant /antioxidant status ratio (/TOS/TAS) as oxidative stress (OS) index (OSI) and BLLPs levels were determined. eNOSP was identified from genomic DNA samples with VNTR analysis. RESULTS There is no significant difference between AG and CG including the genotype groups for NO, eNOS and BLLPs and eNOSP has no role. However, AG's NO (29%, p > .05) and TAS levels were significantly higher (p = .001) than those of CG, whereas OSI (p = .001) and OxLDL (p = .011) values were significantly lower. On the other hand, NO value of the athletic bb group was 29% higher compared with the control and the a carrier (aC = aa + ab) group. CONCLUSIONS These findings suggest that regular aerobic exercise improves blood NO levels and antioxidant capacity, while decreasing OS levels including OxLDL, but not eNOS and BLLPs in the athletes. Although the polymorphism does not have a modifying effect on these effects, bb genotype group may benefit more from training for NO than aC group due to genetic tendency.
Collapse
|
128
|
NejadShahrokhAbadi R, Zangouei AS, Mohebbati R, Shafei MN. Determining the cardiovascular effects of nitric oxide in the dorsolateral Periaqueductal Gray (dlPAG) in anaesthetised rats. J Taibah Univ Med Sci 2020; 15:502-508. [PMID: 33318742 PMCID: PMC7715464 DOI: 10.1016/j.jtumed.2020.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The dorsolateral periaqueductal gray (dlPAG) is an area located in the brain stem that performs a host of functions including cardiovascular regulation. Owing to the presence of nitric oxide (NO) in this area, we investigated its effect on the cardiovascular system. METHODS We divided rats into four groups: 1) control; 2) l-arginine (L-Arg, a precursor for nitric oxide, 60 nmol); 3) l-NAME (N omega-nitro-l-arginine methyl ester, a nitric oxide synthase inhibitor, 90 nmol); and 4) sodium nitroprusside (SNP, a nitric oxide donor, 27 nmol). After anaesthesia, the rats were mounted on a stereotaxic apparatus and the drugs were microinjected into the dlPAG. Cardiovascular parameters were continuously recorded by a PowerLab system connected to the cannulated femoral artery via a pressure transducer. The changes (Δ) of systolic blood pressure (SBP), mean arterial pressure (MAP), and heart rate (HR) were calculated at different times as compared to the control group. RESULTS In the l-NAME group, the ΔSBP, ΔMAP, and ΔHR were not significantly affected compared to the control group. In the L-Arg group, ΔSBP and ΔMAP increased; however, only SBP showed a significant increase compared to the control group. In the SNP group, SBP and MAP were significantly affected in comparison to the controls. Additionally, ΔHR decreased in both L-Arg and SNP, but was only significant in L-Arg. CONCLUSION Our study showed that NO of dlPAG has a pressor effect and attenuates baroreflex bradycardia. However, its pressor effect is more significant.
Collapse
Affiliation(s)
| | - Amir Sadra Zangouei
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Mohebbati
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Naser Shafei
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
129
|
McBride A, Chanh HQ, Fraser JF, Yacoub S, Obonyo NG. Microvascular dysfunction in septic and dengue shock: Pathophysiology and implications for clinical management. Glob Cardiol Sci Pract 2020; 2020:e202029. [PMID: 33447608 PMCID: PMC7773436 DOI: 10.21542/gcsp.2020.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The microcirculation comprising of arterioles, capillaries and post-capillary venules is the terminal vascular network of the systemic circulation. Microvascular homeostasis, comprising of a balance between vasoconstriction, vasodilation and endothelial permeability in healthy states, regulates tissue perfusion. In severe infections, systemic inflammation occurs irrespective of the infecting microorganism(s), resulting in microcirculatory dysregulation and dysfunction, which impairs tissue perfusion and often precedes end-organ failure. The common hallmarks of microvascular dysfunction in both septic shock and dengue shock, are endothelial cell activation, glycocalyx degradation and plasma leak through a disrupted endothelial barrier. Microvascular tone is also impaired by a reduced bioavailability of nitric oxide. In vitro and in vivo studies have however demonstrated that the nature and extent of microvascular dysfunction as well as responses to volume expansion resuscitation differ in these two clinical syndromes. This review compares and contrasts the pathophysiology of microcirculatory dysfunction in septic versus dengue shock and the attendant effects of fluid administration during resuscitation.
Collapse
Affiliation(s)
- Angela McBride
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam.,Brighton and Sussex Medical School, United Kingdom
| | - Ho Q Chanh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
| | - John F Fraser
- Critical Care Research Group, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Sophie Yacoub
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam.,Centre for Tropical Medicine and Global Health, University of Oxford, United Kingdom
| | - Nchafatso G Obonyo
- Critical Care Research Group, Brisbane, Australia.,KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.,Initiative to Develop African Research Leaders, Kilifi, Kenya
| |
Collapse
|
130
|
Jiang W, Liu C, Deng M, Wang F, Ren X, Fan Y, Du J, Wang Y. H 2S promotes developmental brain angiogenesis via the NOS/NO pathway in zebrafish. Stroke Vasc Neurol 2020; 6:244-251. [PMID: 33246971 PMCID: PMC8258041 DOI: 10.1136/svn-2020-000584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/19/2020] [Accepted: 10/28/2020] [Indexed: 01/04/2023] Open
Abstract
Background Hydrogen sulphide (H2S) is considered as the third member of the gasotransmitter family, along with nitric oxide (NO) and carbon monoxide. H2S has been reported to induce angiogenesis by promoting the growth, migration and tube-like structure formation of endothelial cells. Those studies were conducted in conditions of cell culture, mouse Matrigel plug assay model, rat wound healing model or rat hindlimb ischaemia model. Recent in vivo studies showed the physiological importance of H2S in muscle angiogenesis. However, the importance of endogenous H2S for brain angiogenesis during development remains unknown. We therefore aimed at determining the role of H2S in brain vascular development. Methods and results Both knockdown and knockout of H2S-producing enzymes, cystathionine β-synthase (cbs) and cystathionine γ-lyase (cth), using morpholino oligonucleotides and clustered regularly interspaced short palindromic repeats/Cas9-mediated mutation, impaired brain vascular development of larval zebrafish. Incubation with the slow-releasing H2S donor GYY4137 alleviated the defects of brain vascular development in cbs and cth morphants. Quantitative analysis of the midbrain vascular network showed that H2S enhances angiogenesis without affecting the topological structure of the brain vasculature. Mechanically, nitric oxide synthase 2a (nos2a) expression and NO production were decreased in both cbs and cth morphants. Overexpression of nos2a by coinjection of cbs or cth MO with full-length zebrafish nos2a mRNA alleviated the brain vascular developmental defects in cbs and cth morphants. Conclusion We conclude that H2S promotes brain developmental angiogenesis via the NOS/NO pathway in zebrafish.
Collapse
Affiliation(s)
- Weiqing Jiang
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Chen Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingzhu Deng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fei Wang
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Xiao Ren
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Yilin Fan
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yonggang Wang
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
131
|
Wang WL, Ge TY, Chen X, Mao Y, Zhu YZ. Advances in the Protective Mechanism of NO, H 2S, and H 2 in Myocardial Ischemic Injury. Front Cardiovasc Med 2020; 7:588206. [PMID: 33195476 PMCID: PMC7661694 DOI: 10.3389/fcvm.2020.588206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Myocardial ischemic injury is among the top 10 leading causes of death from cardiovascular diseases worldwide. Myocardial ischemia is caused mainly by coronary artery occlusion or obstruction. It usually occurs when the heart is insufficiently perfused, oxygen supply to the myocardium is reduced, and energy metabolism in the myocardium is abnormal. Pathologically, myocardial ischemic injury generates a large number of inflammatory cells, thus inducing a state of oxidative stress. This sharp reduction in the number of normal cells as a result of apoptosis leads to organ and tissue damage, which can be life-threatening. Therefore, effective methods for the treatment of myocardial ischemic injury and clarification of the underlying mechanisms are urgently required. Gaseous signaling molecules, such as NO, H2S, H2, and combined gas donors, have gradually become a focus of research. Gaseous signaling molecules have shown anti-apoptotic, anti-oxidative and anti-inflammatory effects as potential therapeutic agents for myocardial ischemic injury in a large number of studies. In this review, we summarize and discuss the mechanism underlying the protective effect of gaseous signaling molecules on myocardial ischemic injury.
Collapse
Affiliation(s)
| | | | - Xu Chen
- Guilin Medical College, Guilin, China
| | - Yicheng Mao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yi-Zhun Zhu
- Guilin Medical College, Guilin, China.,Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
132
|
Zheng R, da Rosa G, Dans PD, Peluffo RD. Molecular Determinants for Nitric Oxide Regulation of the Murine Cationic Amino Acid Transporter CAT-2A. Biochemistry 2020; 59:4225-4237. [PMID: 33135877 DOI: 10.1021/acs.biochem.0c00729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cationic amino acid transporters (CATs) supply cells with essential and semiessential dibasic amino acids. Among them, l-arginine is the substrate for nitric oxide synthases (NOS) to produce nitric oxide (NO), a key signaling molecule and second messenger. In cardiac preparations, we showed that NO acutely and directly modulates transport activity by noncompetitively inhibiting these CATs. We hypothesize that this NO regulation occurs through modification of cysteine residues in CAT proteins. Homology modeling and a computational chemistry approach identified Cys347 as one of two putative targets for NO binding, of 15 Cys residues present in the low-affinity mouse CAT-2A (mCAT-2A). To test this prediction, mammalian cell lines overexpressing mCAT-2A were used for site-directed mutagenesis and uptake studies. When Cys347 was replaced with alanine (Cys347Ala), mCAT-2A became insensitive to inhibition by NO donors. In addition, the transport capacity of this variant decreased by >50% compared to that of the control, without affecting membrane expression levels or apparent affinities for the transported amino acids. Interestingly, replacing Cys347 with serine (Cys347Ser) restored uptake levels to those of the control while retaining NO insensitivity. Other Cys residues, when replaced with Ala, still produced a NO-sensitive CAT-2A. In cells co-expressing NOS and mCAT-2A, exposure to extracellular l-arginine inhibited the uptake activity of control mCAT-2A, via NO production, but not that of the Cys347Ser variant. Thus, the -SH moiety of Cys347 is largely responsible for mCAT-2A inhibition by NO. Because of the endogenous NO effect, this modulation is likely to be physiologically relevant and a potential intervention point for therapeutics.
Collapse
Affiliation(s)
- Ruifang Zheng
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Avenue, Newark, New Jersey 07103, United States
| | - Gabriela da Rosa
- Laboratory of Molecular Microbiology, DEPBIO, School of Sciences-School of Chemistry, Universidad de la República, 11400 Montevideo, Uruguay.,Functional Genomics Laboratory, Institut Pasteur of Montevideo, Mataojo 2020, CP, 11400 Montevideo, Uruguay.,Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de la República, Rivera 1350, CP, 50000 Salto, Uruguay
| | - Pablo D Dans
- Functional Genomics Laboratory, Institut Pasteur of Montevideo, Mataojo 2020, CP, 11400 Montevideo, Uruguay.,Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de la República, Rivera 1350, CP, 50000 Salto, Uruguay
| | - R Daniel Peluffo
- Department of Pharmacology, Physiology and Neuroscience, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Avenue, Newark, New Jersey 07103, United States.,Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de la República, Rivera 1350, CP, 50000 Salto, Uruguay
| |
Collapse
|
133
|
Kılıç E, Çağlayan B, Caglar Beker M. Physiological and pharmacological roles of melatonin in the pathophysiological components of cellular injury after ischemic stroke. Turk J Med Sci 2020; 50:1655-1664. [PMID: 32962330 PMCID: PMC7672349 DOI: 10.3906/sag-2008-32] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Apart from its metabolic or physiological functions, melatonin has a potent cytoprotective activity in the physiological and pathological conditions. It is synthetized by the pineal gland and released into the blood circulation but particularly cerebrospinal fluid in a circadian manner. It can also easily diffuse through cellular membranes due its small size and lipophilic structure. Its cytoprotective activity has been linked to its potent free radical scavenger activity with the desirable characteristics of a clinically- reliable antioxidant. Melatonin detoxifies oxygen and nitrogen-based free radicals and oxidizing agents, including the highly toxic hydroxyl-and peroxynitrite radicals, initiating cellular damage. However, the cytoprotective activity of melatonin is complex and cannot be solely limited to its free radical scavenger activity. It regulates cellular signaling pathways through receptor– dependent and independent mechanisms. Most of these downstream molecules, such as PI3K/AKT pathway components, also contribute to the cytoprotective effects of melatonin. In this term, melatonin is a promising molecule for the treatment of neurodegenerative disorders, such as ischemic stroke, which melatonin reduces ischemic brain injury in animal models of ischemic stroke. It regulates also circadian rhythm proteins after ischemic stroke, playing roles in cellular survival. In this context, present article summarizes the possible role of melatonin in the pathophysiological events after ischemic stroke.
Collapse
Affiliation(s)
- Ertuğrul Kılıç
- Department of Physiology, School of Medicine, İstanbul Medipol University, İstanbul, Turkey,Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
| | - Berrak Çağlayan
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey,Department of Medical Biology, International School of Medicine, İstanbul Medipol University, İstanbul, Turkey
| | - Mustafa Caglar Beker
- Department of Physiology, School of Medicine, İstanbul Medipol University, İstanbul, Turkey,Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
| |
Collapse
|
134
|
Dong Y, Li XR, Li J, Zang Y, Li X. Selective and sensitive fluorescence imaging reveals microenvironment-dependent behavior of NO modulators in the endothelial system. J Pharm Anal 2020; 10:466-472. [PMID: 33133730 PMCID: PMC7591781 DOI: 10.1016/j.jpha.2020.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/26/2020] [Accepted: 05/22/2020] [Indexed: 11/28/2022] Open
Abstract
Nitric oxide (NO) is a second messenger playing crucial roles in the signaling of a variety of cellular functions. Due to its pathophysiological significance, various NO modulators have been developed to explore NO pathways and some have been used as therapies. These modulators are often used directly to observe pharmacological effects in cell lines, but their actual effect on intracellular NO level is seldom analyzed. Herein, facilitated by a selective and sensitive fluorescence probe, we observed that some NO modulators displayed unexpected behaviors with both NO scavenger carboxy-PTIO and endothelial nitric oxide synthase (eNOS) inhibitor N(ω)-nitro-l-arginine methyl ester (l-NAME) failing to decrease intracellular free NO level in EA. hy926 cells while NO donor diethylamine-NONOate (DEA·NONOate) and eNOS activator calcimycin (A23187) failing to increase free NO level in human umbilical vein endothelial cell line (HUV-EC-C), although the reagents were confirmed to work normally in the primary human umbilical vein endothelial cells (primary HUVECs) and RAW 264.7 macrophage cells. Further research suggested that these unusual behaviors might be attributed to the cellular microenvironments including both the NO synthase (NOS) level and the endogenous glutathione (GSH) level. Genetically manipulating eNOS level in both cells restores the expected response, while decreasing GSH level restores the ability of DEA·NONOate to increase NO level in HUV-EC-C. These results reveal that the cellular microenvironment has a profound impact on pharmacological effect. Our study suggests GSH as a reservoir for NO in live cells and highlights the value of chemical probes as valuable tools to reveal microenvironment-dependent pharmacological effects. An imaging based method for evaluating the efficacy of NO modulators in live cells was developed. Some NO modulators were observed yielding unexpected effects on cellular NO levels in some endothelial cell lines. Further study unveiled intracellular microenvironments have profound effects on the efficacy of NO modulators in live cells. This result highlights the pitfall of the specific cellular microenvironments on drug efficacy.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Rong Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Open Studio for Draggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), 1 Wenhai Road, Aoshanwei, Jimo, Qingdao, 266237, China
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xin Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
135
|
Abdelgawad ME, Darwish H, Nabawy MM, El-mezayen H. Development of novel score based on Angiogenic panel for accurate diagnosis of hepatocellular carcinoma among hepatitis C virus high-risk patients. INFECTION GENETICS AND EVOLUTION 2020; 85:104572. [DOI: 10.1016/j.meegid.2020.104572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
|
136
|
Ghotbeddin Z, Basir Z, Jamshidian J, Delfi F. Modulation of behavioral responses and CA1 neuronal death by nitric oxide in the neonatal rat's hypoxia model. Brain Behav 2020; 10:e01841. [PMID: 32940009 PMCID: PMC7667332 DOI: 10.1002/brb3.1841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/22/2020] [Accepted: 08/29/2020] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION Neonatal hypoxia leads to cognitive and movement impairments that might persist throughout life. Hypoxia impairs hippocampal blood circulation and metabolism. The exact mechanisms underlying hypoxia-induced memory impairment are not fully understood. Nitric oxide (NO) is a key neuromodulator that regulates cerebral blood flow. In this study, we aimed to evaluate the possible role of NO on behavioral and histomorphometric changes in the hippocampus following hypoxia in neonate rats. MATERIAL AND METHODS Neonate male rats (n = 28) were randomly divided into 4 groups: control, hypoxia, hypoxia plus L-NAME (20 mg/kg), and hypoxia plus L-arginine (200 mg/kg). Drugs were injected intraperitoneally for seven consecutive days. Hypoxia was induced by keeping rats in a hypoxic chamber (7% oxygen and 93% nitrogen intensity). Ten to 14 days after hypoxia, behavioral changes were measured using a shuttle box, a rotarod, and an open field test. The histological changes in the hippocampus were measured using H&E and Nissl staining methods. RESULTS Findings showed that hypoxia caused significant atrophy in the hippocampus. Furthermore, the administration of L-NAME decreased the atrophy of the hippocampus in comparison with the hypoxic group. Behavioral results showed that hypoxia impaired memory performance and motor activity responses. Additionally, the administration of L-NAME improved behavioral performance in a significant manner compared with the hypoxic group. CONCLUSIONS Hypoxia damaged the neurons of hippocampal CA1 region and induced memory impairment. The NOS inhibitor, L-NAME, significantly attenuated the negative effects of hypoxia on behavior and observed changes in the hippocampus.
Collapse
Affiliation(s)
- Zohreh Ghotbeddin
- Department of PhysiologyFaculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
- Stem Cell and Transgenic Technology Research CenterShahid Chamran University of AhvazAhvazIran
| | - Zahra Basir
- Department of HistologyFaculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
| | - Javad Jamshidian
- Department of PharmacologyFaculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
| | - Farideh Delfi
- Department of PhysiologyFaculty of Veterinary MedicineShahid Chamran University of AhvazAhvazIran
| |
Collapse
|
137
|
Orsini SS, James KL, Reyes DJ, Couto‐Rodriguez RL, Gulko MK, Witte A, Carroll RK, Rice KC. Bacterial-like nitric oxide synthase in the haloalkaliphilic archaeon Natronomonas pharaonis. Microbiologyopen 2020; 9:e1124. [PMID: 33306280 PMCID: PMC7658456 DOI: 10.1002/mbo3.1124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
Bacterial nitric oxide (NO) synthases (bNOS) play diverse and important roles in microbial physiology, stress resistance, and virulence. Although bacterial and mammalian NOS enzymes have been well-characterized, comparatively little is known about the prevalence and function of NOS enzymes in Archaea. Analysis of archaeal genomes revealed that highly conserved bNOS homologs were restricted to members of the Halobacteria. Of these, Natronomonas pharaonis NOS (npNOS) was chosen for further characterization. NO production was confirmed in heterologously expressed His-tagged npNOS by coupling nitrite production from N-hydroxy-L-arginine in an H2O2-supported reaction. Additionally, the nos gene was successfully targeted and disrupted to create a Nmn. pharaonis nos mutant by adapting an established Natrialba magadii transformation protocol. Genome re-sequencing of this mutant revealed an additional frameshift in a putative cation-acetate symporter gene, which could contribute to altered acetate metabolism in the nos mutant. Inactivation of Nmn. pharaonis nos was also associated with several phenotypes congruent with bacterial nos mutants (altered growth, increased oxygen consumption, increased pigment, increased UV susceptibility), suggesting that NOS function may be conserved between bacteria and archaea. These studies are the first to describe genetic inactivation and characterization of a Nmn. pharaonis gene and provides enhanced tools for probing its physiology.
Collapse
Affiliation(s)
- Silvia S. Orsini
- Department of Microbiology and Cell ScienceIFASUniversity of FloridaGainesvilleFLUSA
- Present address:
Pharma ServicesViral Vector ServicesThermo Fisher ScientificAlachuaFLUSA
| | - Kimberly L. James
- Department of Microbiology and Cell ScienceIFASUniversity of FloridaGainesvilleFLUSA
| | - Destiny J. Reyes
- Department of Microbiology and Cell ScienceIFASUniversity of FloridaGainesvilleFLUSA
- Present address:
Pharma ServicesViral Vector ServicesThermo Fisher ScientificAlachuaFLUSA
| | | | - Miriam K. Gulko
- Department OesterheltMax Planck Institut für BiochemieMartinsriedGermany
| | - Angela Witte
- Department of Microbiology, Immunobiology and GeneticsMPL LaboratoriesUniversity of ViennaViennaAustria
| | | | - Kelly C. Rice
- Department of Microbiology and Cell ScienceIFASUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
138
|
Natural iridoids from Patrinia heterophylla showing anti-inflammatory activities in vitro and in vivo. Bioorg Chem 2020; 104:104331. [PMID: 33142407 DOI: 10.1016/j.bioorg.2020.104331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/13/2020] [Accepted: 09/28/2020] [Indexed: 01/19/2023]
Abstract
Inflammation, especially chronic inflammation, has been found to be closely related to the pathology of many diseases and the discovery of bioactive natural products to inhibit NO production is one of strategies to treat inflammation. In our continuous search for bioactive natural substances as potential anti-inflammatory agents, five new compounds (1-5) were extracted and purified from Patrinia heterophylla. The NMR and MS data analysis, along with electronic circular dichroism (ECD) calculations, led to the identification of these isolates, which were new iridoids. Using cell and zebrafish models, the in vitro and in vivo anti-inflammatory effects were conducted to evaluate the potency of anti-inflammation of these compounds. The preliminary mechanism was explored using molecular docking and Western blotting experiments.
Collapse
|
139
|
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: 1.3] [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
|
140
|
Nakamura N, Pence LM, Cao Z, Beger RD. Distinct lipid signatures are identified in the plasma of rats with chronic inflammation induced by estradiol benzoate and sex hormones. Metabolomics 2020; 16:95. [PMID: 32895772 DOI: 10.1007/s11306-020-01715-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Prostatitis is likely to occur in younger or middle-aged men, while prostate cancer is likely to occur in older men. Although amino acids and lipids as biomarkers of prostate cancer have been examined using prostate cancer cell lines/tissues, no previous studies have evaluated amino acids or lipids as potential chronic prostatitis biomarkers. OBJECTIVES The study's aim was to identify amino acids and lipids that could serve as potential biomarkers of chronic prostatitis. METHODS We profiled the amino acids and lipids found in plasma from rats collected in a previous study. In brief, a total of 148 Sprague-Dawley rats (offspring) were dosed with estradiol benzoate (EB) on postnatal days (PNDs) 1, 3 and 5, and subsequently dosed with testosterone (T)/estradiol (E) tubes via subcutaneous implants from PND 90 to 200. Plasma was collected on PNDs 30, 90, 100, 145 and 200. Analysis was conducted with a Xevo TQ-S triple-quadrupole mass spectrometer using a Biocrates AbsoluteIDQ p180 kit. RESULTS Plasma acylcarnitines [(C2, C16:1, C18, C18:1, C18:1-OH, and C18:2)], glycerophospholipids (lysophosphatidylcholine-acyl, -di-acyl, and -di-acyl acyl-alkyl) and sphingomyelins [SM (OH) C16:1, SM C18:0, SM C18:1, and SM C20:2] significantly increased on PND 145, when chronic inflammation was observed in the dorsolateral prostate of rats dosed with EB, T, and E. No statistical significances of amino acid levels were observed in the EB + T + E group on PND 145. CONCLUSION Exposure to EB, T, and E altered lipid levels in rat plasma with chronic prostate inflammation. These findings suggest that the identified lipids may be predictive chronic prostatitis biomarkers. The results require confirmation through additional nonclinical and human studies.
Collapse
Affiliation(s)
- Noriko Nakamura
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Lisa M Pence
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Zhijun Cao
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Richard D Beger
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| |
Collapse
|
141
|
Darling R, Senapati S, Christiansen J, Liu L, Ramer-Tait AE, Narasimhan B, Wannemuehler M. Polyanhydride Nanoparticles Induce Low Inflammatory Dendritic Cell Activation Resulting in CD8 + T Cell Memory and Delayed Tumor Progression. Int J Nanomedicine 2020; 15:6579-6592. [PMID: 32982219 PMCID: PMC7490050 DOI: 10.2147/ijn.s261041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/02/2020] [Indexed: 12/22/2022] Open
Abstract
Introduction Adjuvants and immunotherapies designed to activate adaptive immunity to eliminate infectious disease and tumors have become an area of interest aimed at providing a safe and effective strategy to prevent or eliminate disease. Existing approaches would benefit from the development of immunization regimens capable of inducing efficacious cell-mediated immunity directed toward CD8+ T cell-specific antigens. This goal is critically dependent upon appropriate activation of antigen-presenting cells (APCs) most notably dendritic cells (DCs). In this regard, polyanhydride particles have been shown to be effectively internalized by APCs and induce activation. Methods Here, a prophylactic vaccine regimen designed as a single-dose polyanhydride nanovaccine encapsulating antigen is evaluated for the induction of CD8+ T cell memory in a model system where antigen-specific protection is restricted to CD8+ T cells. Bone marrow-derived dendritic cells (BMDCs) are used as an in vitro model system to evaluate the magnitude and phenotype of APC activation. Primary DCs, particularly those with described ability to activate CD8+ T cells, are also evaluated for their in vitro responses to polyanhydride nanoparticles. Results Herein, polyanhydride nanoparticles are shown to induce potent in vitro upregulation of costimulatory molecules on the cell surface of BMDCs. In contrast to the classically used TLR agonists, nanoparticles did not induce large amounts of pro-inflammatory cytokines, did not induce characteristic metabolic response of DCs, nor produce innate antimicrobial effector molecules, such as nitric oxide (NO). The polyanhydride nanovaccine results in protective CD8+ T cell responses as measured by inhibition of tumor progression and survival. Discussion Together, these results suggest that the use of a polyanhydride-based nanovaccine can be an effective approach to inducing antigen-specific CD8+ T cell memory by providing antigen delivery and DC activation while avoiding overt inflammatory responses typically associated with traditional adjuvants.
Collapse
Affiliation(s)
- Ross Darling
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, USA
| | - Sujata Senapati
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - John Christiansen
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, USA
| | - Luman Liu
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA.,Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Michael Wannemuehler
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, USA.,Nanovaccine Institute, Iowa State University, Ames, IA, USA
| |
Collapse
|
142
|
Arya VS, Kanthlal SK, Linda G. The role of dietary polyphenols in inflammatory bowel disease: A possible clue on the molecular mechanisms involved in the prevention of immune and inflammatory reactions. J Food Biochem 2020; 44:e13369. [PMID: 32885438 DOI: 10.1111/jfbc.13369] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/08/2020] [Accepted: 06/16/2020] [Indexed: 12/17/2022]
Abstract
Inflammatory bowel disease (IBD) is one of the major complications of the gastrointestinal tract, characterized by chronic inflammation, which disturbs the quality of life of the affected individuals. Genetic predisposition, immune, inflammatory, and enzyme-mediated signaling cascades are the primary mechanisms involved in the pathogenesis of the disease. Currently, the treatment strategy involves the maintenance of remission and induction of inflammation by anti-inflammatory agents and immune suppressants. Polyphenol-containing diets, including fruits and vegetables of regular use, possess anti-inflammatory, and antioxidant potential through the inhibition of major contributing pathways to IBD. This review discusses the role of these dietary polyphenols in downregulating the major signaling cascades in IBD. Our review encourages the development of nutritional strategies to improve the efficiency of current therapies for IBD and reduce the risks of side effects associated with conventional therapy. PRACTICAL APPLICATIONS: At present, almost every third person in society is under stress and having chronic disorders like diabetes, arthritis, allergy, cardiovascular disease, IBD, etc. This insists on the direct/indirect role of changes in the lifestyle for such deterioration in society. This review would emphasize the medicinal value of polyphenols present in fruits and vegetables for chronic inflammatory disorders. This concept portrays the food components which have the potential to promote health, improve general well-being, and reduce the risk of IBD. We propose to add fruits with bioactive polyphenols in the regular diet to help in preventing the immune-mediated intestinal chronic inflammatory syndrome and reduce the risks of colorectal cancer development.
Collapse
Affiliation(s)
- V S Arya
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - S K Kanthlal
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - Geevarghese Linda
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| |
Collapse
|
143
|
Aulak KS, Barnes JW, Tian L, Mellor NE, Haque MM, Willard B, Li L, Comhair SC, Stuehr DJ, Dweik RA. Specific O-GlcNAc modification at Ser-615 modulates eNOS function. Redox Biol 2020; 36:101625. [PMID: 32863226 PMCID: PMC7334407 DOI: 10.1016/j.redox.2020.101625] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/11/2020] [Accepted: 06/24/2020] [Indexed: 01/17/2023] Open
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a progressive and devastating disease characterized by vascular smooth muscle and endothelial cell proliferation leading to a narrowing of the vessels in the lung. The increased resistance in the lung and the higher pressures generated result in right heart failure. Nitric Oxide (NO) deficiency is considered a hallmark of IPAH and altered function of endothelial nitric oxide synthase (eNOS), decreases NO production. We recently demonstrated that glucose dysregulation results in augmented protein serine/threonine hydroxyl-linked N-Acetyl-glucosamine (O-GlcNAc) modification in IPAH. In diabetes, dysregulated glucose metabolism has been shown to regulate eNOS function through inhibition of Ser-1177 phosphorylation. However, the link between O-GlcNAc and eNOS function remains unknown. Here we show that increased protein O-GlcNAc occurs on eNOS in PAH and Ser-615 appears to be a novel site of O-GlcNAc modification resulting in reduced eNOS dimerization. Functional characterization of Ser-615 demonstrated the importance of this residue on the regulation of eNOS activity through control of Ser-1177 phosphorylation. Here we demonstrate a previously unidentified regulatory mechanism of eNOS whereby the O-GlcNAc modification of Ser-615 results in reduced eNOS activity and endothelial dysfunction under conditions of glucose dysregulation.
Collapse
Affiliation(s)
- Kulwant S Aulak
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA
| | - Jarrod W Barnes
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA
| | - Liping Tian
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA
| | - Noel E Mellor
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA
| | - Mohammad M Haque
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA
| | - Belinda Willard
- Mass Spectrometry Laboratory for Protein Sequencing, Cleveland Clinic, OH, USA
| | - Ling Li
- Mass Spectrometry Laboratory for Protein Sequencing, Cleveland Clinic, OH, USA
| | - Suzy C Comhair
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA
| | - Dennis J Stuehr
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA
| | - Raed A Dweik
- Inflammation and Immunity, Lerner Research Institute. Cleveland Clinic, OH, USA; Department of Pulmonary, Allergy and Critical Care Medicine. Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
144
|
Wang RJ, Chen K, Xing LS, Lin Z, Zou Z, Lu Z. Reactive oxygen species and antimicrobial peptides are sequentially produced in silkworm midgut in response to bacterial infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 110:103720. [PMID: 32344046 DOI: 10.1016/j.dci.2020.103720] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 06/11/2023]
Abstract
The silkworm, Bombyx mori, is utilized as a research model in many aspects of biological studies, including genetics, development and immunology. Previous biochemical and genomic studies have elucidated the silkworm immunity in response to infections elicited by bacteria, fungi, microsporidia, and viruses. The intestine serves as the front line in the battle between insects and ingested harmful microorganisms. In this study, we performed RNA sequencing (RNA-seq) of the larval silkworm midgut after oral infection with the Gram-positive bacterium Bacillus bombysepticus and the Gram-negative bacterium Yersinia pseudotuberculosis. This enables us to get a comprehensive understanding of the midgut responses to bacterial infection. We found that B. bombysepticus induced much stronger immune responses than Y. pseudotuberculosis did. Bacterial infection resulted in more energy consumption including carbohydrates and fatty acids. The midgut immune system was characterized by the generation of reactive oxygen species and antimicrobial peptides. The former played a critical role in eliminating invading bacteria during early stage, while the latter executed during late stage. Our results provide an integrated insight into the midgut systematic responses to bacterial infection.
Collapse
Affiliation(s)
- Rui-Juan Wang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China.
| | - Kangkang Chen
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China.
| | - Long-Sheng Xing
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Zhe Lin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, China.
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China; State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
145
|
Transphyletic conservation of nitric oxide synthase regulation in cephalochordates and tunicates. Dev Genes Evol 2020; 230:329-338. [PMID: 32839880 DOI: 10.1007/s00427-020-00668-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 08/16/2020] [Indexed: 12/19/2022]
Abstract
Nitric oxide synthase is ubiquitously present in metazoans and is involved in a wide range of biological processes. Three distinct Nos genes have been so far identified in vertebrates exhibiting a complex expression pattern and transcriptional regulation. Nevertheless, although independent events of Nos duplication have been observed in several taxa, only few studies described the regulatory mechanisms responsible for their activation in non-vertebrate animals. To shed light on the mechanisms underlying neuronal-type Nos expression, we focused on two non-vertebrate chordates: the cephalochordate Branchiostoma lanceolatum and the tunicate Ciona robusta. Here, throughout transphyletic and transgenic approaches, we identified genomic regions in both species acting as Nos functional enhancers during development. In vivo analyses of Nos genomic fragments revealed their ability to recapitulate the endogenous expression territories. Therefore, our results suggest the existence of evolutionary conserved mechanisms responsible for neuronal-type Nos regulation in non-vertebrate chordates. In conclusion, this study paves the way for future characterization of conserved transcriptional logic underlying the expression of neuronal-type Nos genes in chordates.
Collapse
|
146
|
Auta J, Gatta E, Davis JM, Zhang H, Pandey SC, Guidotti A. Essential role for neuronal nitric oxide synthase in acute ethanol-induced motor impairment. Nitric Oxide 2020; 100-101:50-56. [PMID: 32278831 PMCID: PMC7428855 DOI: 10.1016/j.niox.2020.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 11/18/2022]
Abstract
The cerebellum is widely known as a motor structure because it regulates and controls motor learning, coordination, and balance. However, it is also critical for non-motor functions such as cognitive processing, sensory discrimination, addictive behaviors and mental disorders. The cerebellum has the highest relative abundance of neuronal nitric oxide synthase (nNos) and is sensitive to ethanol. Although it has been demonstrated that the interaction of γ-aminobutyric acid (GABA) and nitric oxide (NO) might play an important role in the regulation of ethanol-induced cerebellar ataxia, the molecular mechanisms through which ethanol regulates nNos function to elicit this behavioral effect have not been studied extensively. Here, we investigated the dose-dependent effects of acute ethanol treatment on motor impairment using the rotarod behavioral paradigm and the alterations of nNos mRNA expression in cerebellum, frontal cortex (FC), hippocampus and striatum. We also examined the link between acute ethanol-induced motor impairment and nNos by pharmacological manipulation of nNos function. We found that acute ethanol induced a dose-dependent elevation of ethanol blood levels which was associated with the impairment of motor coordination performance and decreased expression of cerebellar nNos. In contrast, acute ethanol increased nNos expression in FC but did not to change the expression for this enzyme in striatum and hippocampus. The effects of acute ethanol were attenuated by l-arginine, a precursor for NO and potentiated by 7-nitroindazole (7-NI), a selective inhibitor of nNos. Our data suggests that differential regulation of nNos mRNA expression in cerebellum and frontal cortex might be involved in acute ethanol-induced motor impairment.
Collapse
Affiliation(s)
- James Auta
- Center for Alcohol Research in Epigenetics, Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, USA.
| | - Eleonora Gatta
- Center for Alcohol Research in Epigenetics, Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, USA
| | - John M Davis
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago, USA
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Alessandro Guidotti
- Center for Alcohol Research in Epigenetics, Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, USA
| |
Collapse
|
147
|
Yang C, Lim W, Song G. Mediation of oxidative stress toxicity induced by pyrethroid pesticides in fish. Comp Biochem Physiol C Toxicol Pharmacol 2020; 234:108758. [PMID: 32289527 DOI: 10.1016/j.cbpc.2020.108758] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/23/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
Abstract
Organophosphate and organochlorine pesticides are banned in most countries because they cause high toxicity and bioaccumulation in non-target organisms. Pyrethroid pesticides have been applied to agriculture and aquaculture since the 1970s to replace traditional pesticides. However, pyrethroids are approximately 1000 times more toxic to fish than to mammals and birds. Fish-specific organs such as the gills and their late metabolic action against this type of pesticide make fish highly susceptible to the toxicity of pyrethroid pesticides. Oxidative stress plays an important role in the neurological, reproductive, and developmental toxicity caused by pyrethroids. Deltamethrin, cypermethrin, and lambda-cyhalothrin are representative pyrethroid pesticides that induce oxidative stress in tissues such as the gills, liver, and muscles of fish and cause histopathological changes. Although they are observed in low concentrations in aquatic environments such as rivers, lakes, and surface water they induce DNA damage and apoptosis in fish. Pyrethroid pesticides cause ROS-mediated oxidative stress in fish species including carp, tilapia, and trout. They also cause lipid peroxidation and alter the state of DNA, proteins, and lipids in the cells of fish. Moreover, changes in antioxidant enzyme activity following pyrethroid pesticide exposure make fish more susceptible to oxidative stress caused by environmental pollutants. In this review, we examine the occurrence of pyrethroid pesticides in the aquatic environment and oxidative stress-induced toxicity in fish exposed to pyrethroids.
Collapse
Affiliation(s)
- Changwon Yang
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Republic of Korea; Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul 02707, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Republic of Korea; Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
148
|
Hockley A, Berger JI, Palmer AR, Wallace MN. Nitric oxide increases gain in the ventral cochlear nucleus of guinea pigs with tinnitus. Eur J Neurosci 2020; 52:4057-4080. [PMID: 32686192 DOI: 10.1111/ejn.14913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/21/2022]
Abstract
Previous work has led to the hypothesis that, during the production of noise-induced tinnitus, higher levels of nitric oxide (NO), in the ventral cochlear nucleus (VCN), increase the gain applied to a reduced input from the cochlea. To test this hypothesis, we noise-exposed 26 guinea pigs, identified evidence of tinnitus in 12 of them and then compared the effects of an iontophoretically applied NO donor or production inhibitor on VCN single unit activity. We confirmed that the mean driven firing rate for the tinnitus and control groups was the same while it had fallen in the non-tinnitus group. By contrast, the mean spontaneous rate had increased for the tinnitus group relative to the control group, while it remained the same for the non-tinnitus group. A greater proportion of units responded to exogenously applied NO in the tinnitus (56%) and non-tinnitus groups (71%) than a control population (24%). In the tinnitus group, endogenous NO facilitated the driven firing rate in 37% (7/19) of neurons and appeared to bring the mean driven rate back up to control levels by a mechanism involving N-methyl-D-aspartic acid (NMDA) receptors. By contrast, in the non-tinnitus group, endogenous NO only facilitated the driven firing rate in 5% (1/22) of neurons and there was no facilitation of driven rate in the control group. The effects of endogenous NO on spontaneous activity were unclear. These results suggest that NO is involved in increasing the gain applied to driven activity, but other factors are also involved in the increase in spontaneous activity.
Collapse
Affiliation(s)
- Adam Hockley
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,School of Life Sciences, University of Nottingham, Nottingham, UK.,Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, USA
| | - Joel I Berger
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| | - Alan R Palmer
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,Hearing Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Mark N Wallace
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, UK.,Hearing Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
149
|
Wright WS, Eshaq RS, Lee M, Kaur G, Harris NR. Retinal Physiology and Circulation: Effect of Diabetes. Compr Physiol 2020; 10:933-974. [PMID: 32941691 PMCID: PMC10088460 DOI: 10.1002/cphy.c190021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In this article, we present a discussion of diabetes and its complications, including the macrovascular and microvascular effects, with the latter of consequence to the retina. We will discuss the anatomy and physiology of the retina, including aspects of metabolism and mechanisms of oxygenation, with the latter accomplished via a combination of the retinal and choroidal blood circulations. Both of these vasculatures are altered in diabetes, with the retinal circulation intimately involved in the pathology of diabetic retinopathy. The later stages of diabetic retinopathy involve poorly controlled angiogenesis that is of great concern, but in our discussion, we will focus more on several alterations in the retinal circulation occurring earlier in the progression of disease, including reductions in blood flow and a possible redistribution of perfusion that may leave some areas of the retina ischemic and hypoxic. Finally, we include in this article a more recent area of investigation regarding the diabetic retinal vasculature, that is, the alterations to the endothelial surface layer that normally plays a vital role in maintaining physiological functions. © 2020 American Physiological Society. Compr Physiol 10:933-974, 2020.
Collapse
Affiliation(s)
- William S Wright
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, South Carolina, USA
| | - Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Minsup Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Gaganpreet Kaur
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
150
|
Haselden WD, Kedarasetti RT, Drew PJ. Spatial and temporal patterns of nitric oxide diffusion and degradation drive emergent cerebrovascular dynamics. PLoS Comput Biol 2020; 16:e1008069. [PMID: 32716940 PMCID: PMC7410342 DOI: 10.1371/journal.pcbi.1008069] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 08/06/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a gaseous signaling molecule that plays an important role in neurovascular coupling. NO produced by neurons diffuses into the smooth muscle surrounding cerebral arterioles, driving vasodilation. However, the rate of NO degradation in hemoglobin is orders of magnitude higher than in brain tissue, though how this might impact NO signaling dynamics is not completely understood. We used simulations to investigate how the spatial and temporal patterns of NO generation and degradation impacted dilation of a penetrating arteriole in cortex. We found that the spatial location of NO production and the size of the vessel both played an important role in determining its responsiveness to NO. The much higher rate of NO degradation and scavenging of NO in the blood relative to the tissue drove emergent vascular dynamics. Large vasodilation events could be followed by post-stimulus constrictions driven by the increased degradation of NO by the blood, and vasomotion-like 0.1-0.3 Hz oscillations could also be generated. We found that these dynamics could be enhanced by elevation of free hemoglobin in the plasma, which occurs in diseases such as malaria and sickle cell anemia, or following blood transfusions. Finally, we show that changes in blood flow during hypoxia or hyperoxia could be explained by altered NO degradation in the parenchyma. Our simulations suggest that many common vascular dynamics may be emergent phenomena generated by NO degradation by the blood or parenchyma.
Collapse
Affiliation(s)
- William Davis Haselden
- Neuroscience Graduate Program, MD/PhD Medical Scientist Training Program, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Ravi Teja Kedarasetti
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Patrick J. Drew
- Neuroscience Graduate Program, MD/PhD Medical Scientist Training Program, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Departments of Biomedical Engineering and Neurosurgery, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|