1
|
Wang Z, Ao J, Lai X, Liu G, Meng Y, Zhang Y, Shi J, Wu Y, Wang C. Decreasing the aggregation of photosensitizers to facilitate energy transfer for improved photodynamic therapy. NANOSCALE 2025; 17:5707-5714. [PMID: 39871776 DOI: 10.1039/d4nr04593a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
The mode of energy transfer between photosensitizers and oxygen determines the yield of singlet oxygen (1O2), crucial for photodynamic therapy (PDT). However, the aggregation of photosensitizers promotes electron transfer while inhibiting pure energy transfer, resulting in the generation of the hypotoxic superoxide anion (O2-) and consumption of substantial oxygen. Herein, we achieve the reduction of the aggregation of photosensitizers to inhibit electron transfer through classical chemical crosslinking, thereby boosting the production of 1O2. Specifically, we constructed a cross-linked hydrogel-like nanophotosensitizer (HA-TPP NHs) via amidation reactions between hyaluronic acid (HA) and tetrakis(4-aminophenyl)porphyrin (TATPP). In HA-TPP NHs, porphyrin is anchored at the crosslinking sites, preventing their close proximity. Simultaneously, HA-TPP NHs swell in a physiological environment due to water absorption, further increasing the distance between porphyrin molecules to avoid their aggregation. Compared to porphyrin-hyaluronic acid assembling nanoparticles (HA-TPP NPs), we find that the 1O2 generation efficiency of HA-TPP NHs is elevated by over 80%. Furthermore, leveraging the targeting capabilities of hyaluronic acid, HA-TPP NHs demonstrate a remarkable anticancer effect in in vitro and in vivo experiments. This study offers a novel insight and method for improving the therapeutic efficacy of PDT.
Collapse
Affiliation(s)
- Zhenhua Wang
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Jianyang Ao
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Xiaoran Lai
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Gang Liu
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Yun Meng
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Yurong Zhang
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Jieyun Shi
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Yelin Wu
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| | - Chaochao Wang
- Institute of Hepatobiliary and Pancreatic Surgery, Department of Hepatobiliary and Pancreatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, P. R. China.
| |
Collapse
|
2
|
Fanfarillo F, Ferraguti G, Lucarelli M, Francati S, Barbato C, Minni A, Ceccanti M, Tarani L, Petrella C, Fiore M. The Impact of ROS and NGF in the Gliomagenesis and their Emerging Implications in the Glioma Treatment. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:449-462. [PMID: 37016521 DOI: 10.2174/1871527322666230403105438] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 04/06/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules derived from molecular oxygen (O2). ROS sources can be endogenous, such as cellular organelles and inflammatory cells, or exogenous, such as ionizing radiation, alcohol, food, tobacco, chemotherapeutical agents and infectious agents. Oxidative stress results in damage of several cellular structures (lipids, proteins, lipoproteins, and DNA) and is implicated in various disease states such as atherosclerosis, diabetes, cancer, neurodegeneration, and aging. A large body of studies showed that ROS plays an important role in carcinogenesis. Indeed, increased production of ROS causes accumulation in DNA damage leading to tumorigenesis. Various investigations demonstrated the involvement of ROS in gliomagenesis. The most common type of primary intracranial tumor in adults is represented by glioma. Furthermore, there is growing attention on the role of the Nerve Growth Factor (NGF) in brain tumor pathogenesis. NGF is a growth factor belonging to the family of neurotrophins. It is involved in neuronal differentiation, proliferation and survival. Studies were conducted to investigate NGF pathogenesis's role as a pro- or anti-tumoral factor in brain tumors. It has been observed that NGF can induce both differentiation and proliferation in cells. The involvement of NGF in the pathogenesis of brain tumors leads to the hypothesis of a possible implication of NGF in new therapeutic strategies. Recent studies have focused on the role of neurotrophin receptors as potential targets in glioma therapy. This review provides an updated overview of the role of ROS and NGF in gliomagenesis and their emerging role in glioma treatment.
Collapse
Affiliation(s)
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Antonio Minni
- Department of Sensory Organs, Sapienza University of Rome, Rome, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell'Alcolismo e le sue Complicanze, Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| |
Collapse
|
3
|
Flores AE, Pascotini ET, Kegler A, Broetto N, Gabbi P, Duarte T, Prado ALC, Duarte MMMF, da Cruz IBM, Dos Santos ARS, Royes LFF, Fighera MR. Worst spasticity in patients post-stroke associated with MNSOD ALA16VAL polymorphism and interleukin-1β. Gene X 2022; 847:146880. [PMID: 36100117 DOI: 10.1016/j.gene.2022.146880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/23/2022] [Accepted: 09/05/2022] [Indexed: 11/04/2022] Open
Abstract
The MnSOD Ala16Val single nucleotide polymorphism (SNP) has shown to be associated to risk factors of several metabolic and vascular diseases. However, little is known about interaction between MnSOD Ala16Val SNP in stroke, a frequent neurologic disease that involves clinic manifestations such as motor deficits and spasticity. In this sense, we decided to investigate the relationship between MnSOD Ala16Val SNP with spasticity in stroke and also its influence on interleukin levels, BDNF, and glycolipid parameters. Eighty post-stroke subjects and 80 healthy controls were investigated. We showed a higher spasticity, levels of total cholesterol, LDL, IL-1β, IL-6, and INF-γ in VV post-stroke group. Interesting, we found a correlation between IL-1β levels and spasticity in VV post-stroke. Triglycerides, glucose levels and caspases (1 and 3) activation were significantly higher, as well as BDNF levels were lower in VV and AV post-stroke. DNA damage was higher in post-stroke group. Thus, we can suggest that the V allele has a worse glycolipid profile, which would facilitate changes in neurovascular homeostasis. These events associated with an increase in inflammatory markers and a reduction in BDNF can contribute with the stroke and a worse clinical evolution in relation to spasticity in patients with VV genotype.
Collapse
Affiliation(s)
- Ariane Ethur Flores
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Eduardo Tanuri Pascotini
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Aline Kegler
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Núbia Broetto
- Curso de Fisioterapia, Departamento de Ginástica e Saúde, Universidade Federal de Pelotas, RS, Brazil
| | - Patricia Gabbi
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Thiago Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | | | - Marta M M F Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Ivana B M da Cruz
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | | | - Luiz Fernando Freire Royes
- Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
4
|
Chen W, Yasen M, Wang H, Zhuang C, Wang Z, Lu S, Jiang L, Lin H. Celecoxib activates autophagy by inhibiting the mTOR signaling pathway and prevents apoptosis in nucleus pulposus cells. BMC Pharmacol Toxicol 2022; 23:90. [PMID: 36457130 PMCID: PMC9714067 DOI: 10.1186/s40360-022-00633-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Intervertebral disc degeneration results from a variety of etiologies, including inflammation and aging. Degenerated intervertebral discs feature down-regulated extracellular matrix synthesis, resulting in losing their ability to retain water and absorb compression. Celecoxib is a well-known selective cyclooxygenase-2 inhibitor for treating arthritis and relieving pain. Nevertheless, the mechanism of Celecoxib for treating inflammation-related intervertebral disc degeneration has not yet been clarified. METHOD Protein synthesis was analyzed by western blot. Fluorescent probes DCFH-DA and MitoSox Red detected reactive oxygen species and were measured by flow cytometry. The activity of the kinase pathway was evaluated by protein phosphorylation. Autophagy was monitored by mRFP-GFP-LC3 transfection and LC3 analysis. Mitochondrial apoptotic proteins were analyzed by western blot and cell membrane integrity was measured by flow cytometry. The autophagic gene was silenced by siRNA. RESULTS In this study, interleukin-1β stimulation reduced the synthesis of aggrecan, type I and II collagen and caused excessive production of reactive oxygen species. We looked for a therapeutic window of Celecoxib for nucleus pulposus cells to regain extracellular matrix synthesis and reduce oxidative stress. To look into nucleus pulposus cells in response to stimuli, enhancement of autophagy was achieved by Celecoxib, confirmed by mRFP-GFP-LC3 transfection and LC3 analysis. The mammalian target of rapamycin and a panel of downstream proteins responded to Celecoxib and propelled autophagy machinery to stabilize homeostasis. Ultimately, inhibition of autophagy by silencing autophagy protein 5 disrupted the protective effects of Celecoxib, culminating in apoptosis. CONCLUSION In summary, we have demonstrated a new use for the old drug Celecoxib that treats intervertebral disc degeneration by enhancing autophagy in nucleus pulposus cells and opening a door for treating other degenerative diseases.
Collapse
Affiliation(s)
- Weisin Chen
- grid.413087.90000 0004 1755 3939Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032 Shanghai, China
| | - Miersalijiang Yasen
- grid.413087.90000 0004 1755 3939Department of Orthopedic Surgery, Zhongshan Hospital Xiamen Branch, Fudan University, 668 Jinhu Rd, District of Huli, Fujian 361015 Xiamen, China
| | - Hanquan Wang
- grid.413087.90000 0004 1755 3939Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032 Shanghai, China
| | - Chenyang Zhuang
- grid.413087.90000 0004 1755 3939Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032 Shanghai, China ,grid.8547.e0000 0001 0125 2443Department of Orthopaedics, Shanghai Geriatric Medical Centre, Fudan University, Shanghai, China
| | - Zixiang Wang
- grid.413087.90000 0004 1755 3939Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032 Shanghai, China
| | - Shunyi Lu
- grid.413087.90000 0004 1755 3939Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032 Shanghai, China
| | - Libo Jiang
- grid.413087.90000 0004 1755 3939Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032 Shanghai, China
| | - Hong Lin
- grid.413087.90000 0004 1755 3939Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032 Shanghai, China ,grid.8547.e0000 0001 0125 2443Department of Orthopaedics, Shanghai Geriatric Medical Centre, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Chen W, Yasen M, Wang H, Zhuang C, Wang Z, Lu S, Jiang L, Lin H. Celecoxib activates autophagy by inhibiting the mTOR signaling pathway and prevents apoptosis in nucleus pulposus cells. BMC Pharmacol Toxicol 2022. [PMID: 36457130 DOI: 10.1186/s40360-022-00633-y/figures/6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Intervertebral disc degeneration results from a variety of etiologies, including inflammation and aging. Degenerated intervertebral discs feature down-regulated extracellular matrix synthesis, resulting in losing their ability to retain water and absorb compression. Celecoxib is a well-known selective cyclooxygenase-2 inhibitor for treating arthritis and relieving pain. Nevertheless, the mechanism of Celecoxib for treating inflammation-related intervertebral disc degeneration has not yet been clarified. METHOD Protein synthesis was analyzed by western blot. Fluorescent probes DCFH-DA and MitoSox Red detected reactive oxygen species and were measured by flow cytometry. The activity of the kinase pathway was evaluated by protein phosphorylation. Autophagy was monitored by mRFP-GFP-LC3 transfection and LC3 analysis. Mitochondrial apoptotic proteins were analyzed by western blot and cell membrane integrity was measured by flow cytometry. The autophagic gene was silenced by siRNA. RESULTS In this study, interleukin-1β stimulation reduced the synthesis of aggrecan, type I and II collagen and caused excessive production of reactive oxygen species. We looked for a therapeutic window of Celecoxib for nucleus pulposus cells to regain extracellular matrix synthesis and reduce oxidative stress. To look into nucleus pulposus cells in response to stimuli, enhancement of autophagy was achieved by Celecoxib, confirmed by mRFP-GFP-LC3 transfection and LC3 analysis. The mammalian target of rapamycin and a panel of downstream proteins responded to Celecoxib and propelled autophagy machinery to stabilize homeostasis. Ultimately, inhibition of autophagy by silencing autophagy protein 5 disrupted the protective effects of Celecoxib, culminating in apoptosis. CONCLUSION In summary, we have demonstrated a new use for the old drug Celecoxib that treats intervertebral disc degeneration by enhancing autophagy in nucleus pulposus cells and opening a door for treating other degenerative diseases.
Collapse
Affiliation(s)
- Weisin Chen
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032, Shanghai, China
| | - Miersalijiang Yasen
- Department of Orthopedic Surgery, Zhongshan Hospital Xiamen Branch, Fudan University, 668 Jinhu Rd, District of Huli, Fujian, 361015, Xiamen, China
| | - Hanquan Wang
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032, Shanghai, China
| | - Chenyang Zhuang
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032, Shanghai, China
- Department of Orthopaedics, Shanghai Geriatric Medical Centre, Fudan University, Shanghai, China
| | - Zixiang Wang
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032, Shanghai, China
| | - Shunyi Lu
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032, Shanghai, China
| | - Libo Jiang
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032, Shanghai, China.
| | - Hong Lin
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road/1609 Xietu Road, 200032, Shanghai, China.
- Department of Orthopaedics, Shanghai Geriatric Medical Centre, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Fois SS, Canu S, Fois AG. The Role of Oxidative Stress in Sarcoidosis. Int J Mol Sci 2021; 22:ijms222111712. [PMID: 34769145 PMCID: PMC8584035 DOI: 10.3390/ijms222111712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 01/15/2023] Open
Abstract
Sarcoidosis is a rare, systemic inflammatory disease whose diagnosis and management can pose a challenge for clinicians and specialists. Scientific knowledge on the molecular pathways that drive its development is still lacking, with no standardized therapies available and insufficient strategies to predict patient outcome. In recent years, oxidative stress has been highlighted as an important factor in the pathogenesis of sarcoidosis, involving several enzymes and molecules in the mechanism of the disease. This review presents current data on the role of oxidative stress in sarcoidosis and its interaction with inflammation, as well as the application of antioxidative therapy in the disease.
Collapse
Affiliation(s)
- Sara Solveig Fois
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy;
- Correspondence:
| | - Sara Canu
- Respiratory Diseases Operative Unit, University Hospital of Sassari, 07100 Sassari, Italy;
| | - Alessandro Giuseppe Fois
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy;
| |
Collapse
|
7
|
Li C, Sun T, Jiang C. Recent advances in nanomedicines for the treatment of ischemic stroke. Acta Pharm Sin B 2021; 11:1767-1788. [PMID: 34386320 PMCID: PMC8343119 DOI: 10.1016/j.apsb.2020.11.019] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/27/2020] [Accepted: 09/13/2020] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is a cerebrovascular disease normally caused by interrupted blood supply to the brain. Ischemia would initiate the cascade reaction consisted of multiple biochemical events in the damaged areas of the brain, where the ischemic cascade eventually leads to cell death and brain infarction. Extensive researches focusing on different stages of the cascade reaction have been conducted with the aim of curing ischemic stroke. However, traditional treatment methods based on antithrombotic therapy and neuroprotective therapy are greatly limited for their poor safety and treatment efficacy. Nanomedicine provides new possibilities for treating stroke as they could improve the pharmacokinetic behavior of drugs in vivo, achieve effective drug accumulation at the target site, enhance the therapeutic effect and meanwhile reduce the side effect. In this review, we comprehensively describe the pathophysiology of stroke, traditional treatment strategies and emerging nanomedicines, summarize the barriers and methods for transporting nanomedicine to the lesions, and illustrate the latest progress of nanomedicine in treating ischemic stroke, with a view to providing a new feasible path for the treatment of cerebral ischemia.
Collapse
Key Words
- AEPO, asialo-erythropoietin
- APOE, apolipoprotein E
- BBB, blood‒brain barrier
- BCECs, brain capillary endothelial cells
- Blood‒brain barrier
- CAT, catalase
- COX-1, cyclooxygenase-1
- CXCR-4, C-X-C chemokine receptor type 4
- Ce-NPs, ceria nanoparticles
- CsA, cyclosporine A
- DAMPs, damage-associated molecular patterns
- GFs, growth factors
- GPIIb/IIIa, glycoprotein IIb/IIIa
- HMGB1, high mobility group protein B1
- Hb, hemoglobin
- ICAM-1, intercellular adhesion molecule-1
- IL-1β, interleukin-1β
- IL-6, interleukin-6
- Ischemic cascade
- LFA-1, lymphocyte function-associated antigen-1
- LHb, liposomal Hb
- MCAO, middle cerebral artery occlusion
- MMPs, matrix metalloproteinases
- MSC, mesenchymal stem cell
- NF-κB, nuclear factor-κB
- NGF, nerve growth factor
- NMDAR, N-methyl-d-aspartate receptor
- NOS, nitric oxide synthase
- NPs, nanoparticles
- NSCs, neural stem cells
- Nanomedicine
- Neuroprotectant
- PBCA, poly-butylcyanoacrylate
- PCMS, poly (chloromethylstyrene)
- PEG, poly-ethylene-glycol
- PEG-PLA, poly (ethylene-glycol)-b-poly (lactide)
- PLGA NPs, poly (l-lactide-co-glycolide) nanoparticles
- PSD-95, postsynaptic density protein-95
- PSGL-1, P-selectin glycoprotein ligand-1
- RBCs, red blood cells
- RES, reticuloendothelial system
- RGD, Arg-Gly-Asp
- ROS, reactive oxygen species
- Reperfusion
- SDF-1, stromal cell-derived factor-1
- SHp, stroke homing peptide
- SOD, superoxide dismutase
- SUR1-TRPM4, sulfonylurea receptor 1-transient receptor potential melastatin-4
- Stroke
- TEMPO, 2,2,6,6-tetramethylpiperidine-1-oxyl
- TIA, transient ischemic attack
- TNF-α, tumor necrosis factor-α
- Thrombolytics
- cRGD, cyclic Arg-Gly-Asp
- e-PAM-R, arginine-poly-amidoamine ester
- iNOS, inducible nitric oxide synthase
- miRNAs, microRNAs
- nNOS, neuron nitric oxide synthase
- siRNA, small interfering RNA
Collapse
|
8
|
Wu XM, Qian C, Jiang F, Bao YX, Qian ZM, Ke Y. The involvement of nuclear factor-κB in astroprotection against ischemia-reperfusion injury by ischemia-preconditioned neurons. J Cell Physiol 2021; 236:4515-4527. [PMID: 33442879 DOI: 10.1002/jcp.30168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/21/2020] [Accepted: 11/06/2020] [Indexed: 11/12/2022]
Abstract
Ischemic preconditioned (IP) neurons protect astrocytes against ischemia/reperfusion (I/R)-induced injury by inhibiting oxidative stress. However, the relevant mechanisms are unknown. Based on the role of nuclear factor-κB (NF-κB) in cell survival and adaption to oxidative stress, we hypothesized that NF-κB might be associated with astroprotection induced by IP neurons via upregulation of antioxidant enzymes. Here, we investigated the effects of IP neurons on NF-κB activation, cell viability, reactive oxygen species (ROS), expression of antioxidant enzymes, erythropoietin (EPO), and tumor necrosis factor α (TNF-α), in the presence or absence of BAY11-7082 (an NF-κB inhibitor), anti-EPO, and anti-TNF-α antibodies, in astrocytes treated with or without I/R. We found that IP neurons could keep NF-κB activation at a relatively higher but beneficial level, and in turn, upregulated the activity of antioxidant enzymes and hence enhanced cell viability and reduced ROS in I/R treated astrocytes. The results collectively indicated that IP neurons are able to significantly inhibit the I/R-induced NF-κB overactivation, probably via EPO and TNF-α, being essential for IP neuron-induced astroprotection under the conditions of I/R. We concluded that NF-κB-mediated antioxidative stress is one of the mechanisms by which IP neurons protect astrocytes against I/R injury.
Collapse
Affiliation(s)
- Xiao-Mei Wu
- Institute of Translational & Precision Medicine and Institute for Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Fei Jiang
- Institute of Translational & Precision Medicine and Institute for Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Yu-Xin Bao
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhong-Ming Qian
- Institute of Translational & Precision Medicine and Institute for Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
- Laboratory of Neuropharmacology, School of Pharmacy & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| |
Collapse
|
9
|
Manganese superoxide dismutase induced by lipoteichoic acid isolated from Staphylococcus aureus regulates cytokine production in THP-1 cells. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2021; 55:36-43. [PMID: 33558048 DOI: 10.1016/j.jmii.2020.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/08/2020] [Accepted: 12/24/2020] [Indexed: 11/23/2022]
Abstract
Lipoteichoic acid isolated from Staphylococcus aureus (aLTA) is known to regulate the production of pro-inflammatory cytokines through TLR2-mediated signaling pathways. In our previous study, we found that aLTA significantly increased manganese superoxide dismutase (MnSOD) in the THP-1 human monocyte-like cell line, but the role of MnSOD in the regulation of cytokine production was not elucidated. In the current study, we found that MnSOD was involved in aLTA-mediated cytokine production. The signaling pathways associated with aLTA-mediated MnSOD induction in THP-1 cells included TLR2-MyD88-IRAK2, JNK (c-Jun N-terminal kinases)1/2 and nuclear factor- κB (NF-κB). We also found MnSOD was involved in the regulation of IL-1β and TNF-α, which were induced by early signaling pathways, including JNK1/2, p38, and NF-κB p65. In addition, MnSOD was also involved in the production of IL-6 and CCL2 in aLTA-stimulated THP-1 cells through activation of late signaling pathways such as JAK2-STAT3. Taken together, our data suggest that aLTA-mediated MnSOD production involved in the regulation of cytokine production and it may be the cause of one of the excessive inflammatory reactions caused by S. aureus.
Collapse
|
10
|
Arend J, Kegler A, Caprara ALF, Gabbi P, Pascotini ET, de Freitas LAV, Duarte MMMF, Broetto N, Furian AF, Oliveira MS, Royes LFF, Fighera MR. MnSOD Ala16Val polymorphism in cognitive dysfunction in patients with epilepsy: A relationship with oxidative and inflammatory markers. Epilepsy Behav 2020; 112:107346. [PMID: 32889510 DOI: 10.1016/j.yebeh.2020.107346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The objective of the study was to evaluate the neurocognitive profile and its relation with Ala16ValMnSOD polymorphism in epilepsy and if these clinical parameters are linked to oxidative stress and inflammatory markers. METHODS Patients with epilepsy (n = 31) and healthy subjects (n = 42) were recruited. A neuropsychological evaluation was performed in both groups through a battery of cognitive tests. Oxidative stress, inflammatory markers, apoptotic factors, and deoxyribonucleic acid (DNA) damage were measured in blood samples. RESULTS Statistical analyses showed the association of MnSOD Ala16Val polymorphism with cognitive impairment, including praxis, perception, attention, language, executive functions, long-term semantic memory, short-term visual memory, and total memory in patients with epilepsy and Valine-Valine (VV) genotype compared with the control group. Compared with the controls and patients with epilepsy, Alanine-Alanine (AA), and Alanine-Valine (AV) genotype, the patients with epilepsy and VV genotype exhibited higher levels of tumor necrosis factor alpha (TNF-α), interleukin 1β (IL-1β), interleukin 6 (IL-6), activation of caspases 1 and 3 (CASP-1 and -3), and DNA damage. Our findings also showed higher carbonyl protein and thiobarbituric acid reactive substances (TBARS) levels as well as an increased superoxide dismutase (SOD) and acetylcholinesterase (AChE) activities in patients with epilepsy and VV genotype. CONCLUSION This study supports the evidence of a distinct neuropsychological profile in patients with epilepsy, especially those with the VV genotype. Furthermore, our results suggest that oxidative and inflammatory pathways may be associated with genetic polymorphism and cognitive dysfunction in patients with epilepsy.
Collapse
Affiliation(s)
- Josi Arend
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Aline Kegler
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil
| | - Ana Letícia Fornari Caprara
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil
| | - Patricia Gabbi
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil
| | - Eduardo T Pascotini
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil
| | - Lori Ane Vargas de Freitas
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil
| | - Marta M M F Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Núbia Broetto
- Centro de Educação Física e Desportos, Departamento de Métodos e Técnicas Desportivas, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil
| | - Ana Flavia Furian
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Mauro Schneider Oliveira
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Luiz Fernando Freire Royes
- Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Departamento de Métodos e Técnicas Desportivas, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Departamento de Métodos e Técnicas Desportivas, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
11
|
Anonna SN, Ahamed SK, Uddin MG, Adnan MT, Uddin SMN, Hussain MS, Millat MS, Bulbul L, Bhatta R, Sarwar MS, Rashid MMU, Chowdhury JA, Islam MS. A clinical evaluation of the alterations in the level of serum zinc, copper, iron, and manganese in the ischemic heart disease patients of Bangladesh - A case-control study. Heliyon 2020; 6:e05311. [PMID: 33102882 PMCID: PMC7578678 DOI: 10.1016/j.heliyon.2020.e05311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/16/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Background Ischemic heart disease (IHD) is a major cause of death globally. Countries vary in their rates, and changes have occurred over time. Nowadays, developing countries pose new public health challenges. Objectives The objective of the present study was to appraise the alterations in the levels of serum Zn, Cu, Fe, and Mn that occur in patients with ischemic heart disease and to depict the correlations of the effects of these changes that lead to the pathogenesis of IHD. Methods Zn, Cu, Fe, and Mn in the IHD patients were determined by Atomic Absorption Spectroscopy (AAS). Results This study evaluated 52 patients with IHD, and 61 healthy volunteers served as controls. The primary outcomes of interest were explored regarding the correlations of the serum levels of these trace elements in patients with IHD. The secondary outcomes were explored in terms of inter-element relations to connect them with the pathogenesis of IHD. Our study found significantly reduced levels of Zn and Cu (2.50 ± 0.19 mg/L and 2.52 ± 0.17 mg/L, respectively) and an elevated level of Fe (148.97 ± 17.25 mg/L) in the patient group with IHD. The level of Mn (7.32 ± 1.23 mg/L) was elevated in the sera of the patients with ischemic heart disease (IHD) compared to healthy control subjects. Conclusion Our results indicate strong associations of the pathogenesis of IHD with depleted serum levels of Zn and Cu and elevated Fe and Mn levels, which may provide a prognostic tool for the treatment of this concerning the disease.
Collapse
Affiliation(s)
- Shamima Nasrin Anonna
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | | | - Md Giash Uddin
- Department of Pharmacy, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Tarek Adnan
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - S M Naim Uddin
- Department of Pharmacy, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Saddam Hussain
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Md Shalahuddin Millat
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Latifa Bulbul
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Rita Bhatta
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Md Mamun Ur Rashid
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Jakir Ahmed Chowdhury
- Department of Pharmaceutical Technology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Mohammad Safiqul Islam
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| |
Collapse
|
12
|
He M, He M, Zhang J, Liu C, Pan Q, Yi J, Chen T. A spatial-confinement hairpin cascade reaction-based DNA tetrahedral amplifier for mRNA imaging in live cells. Talanta 2020; 207:120287. [DOI: 10.1016/j.talanta.2019.120287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 01/05/2023]
|
13
|
Ajmone-Cat MA, Spinello C, Valenti D, Franchi F, Macrì S, Vacca RA, Laviola G. Brain-Immune Alterations and Mitochondrial Dysfunctions in a Mouse Model of Paediatric Autoimmune Disorder Associated with Streptococcus: Exacerbation by Chronic Psychosocial Stress. J Clin Med 2019; 8:1514. [PMID: 31547098 PMCID: PMC6833026 DOI: 10.3390/jcm8101514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022] Open
Abstract
Adverse psychosocial experiences have been shown to modulate individual responses to immune challenges and affect mitochondrial functions. The aim of this study was to investigate inflammation and immune responses as well as mitochondrial bioenergetics in an experimental model of Paediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus (PANDAS). Starting in adolescence (postnatal day 28), male SJL/J mice were exposed to five injections (interspaced by two weeks) with Group-A beta-haemolytic streptococcus (GAS) homogenate. Mice were exposed to chronic psychosocial stress, in the form of protracted visual exposure to an aggressive conspecific, for four weeks. Our results indicate that psychosocial stress exacerbated individual response to GAS administrations whereby mice exposed to both treatments exhibited altered cytokine and immune-related enzyme expression in the hippocampus and hypothalamus. Additionally, they showed impaired mitochondrial respiratory chain complexes IV and V, and reduced adenosine triphosphate (ATP) production by mitochondria and ATP content. These brain abnormalities, observed in GAS-Stress mice, were associated with blunted titers of plasma corticosterone. Present data support the hypothesis that challenging environmental conditions, in terms of chronic psychosocial stress, may exacerbate the long-term consequences of exposure to GAS processes through the promotion of central immunomodulatory and oxidative stress.
Collapse
Affiliation(s)
- Maria Antonietta Ajmone-Cat
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena, 299, I-00161 Rome, Italy.
| | - Chiara Spinello
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, I-00161 Rome, Italy.
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Via Giovanni Amendola 122/O - 70126 Bari, Italy.
| | - Francesca Franchi
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, I-00161 Rome, Italy.
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, I-00161 Rome, Italy.
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Via Giovanni Amendola 122/O - 70126 Bari, Italy.
| | - Giovanni Laviola
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, I-00161 Rome, Italy.
| |
Collapse
|
14
|
Mitra S, Nguyen LN, Akter M, Park G, Choi EH, Kaushik NK. Impact of ROS Generated by Chemical, Physical, and Plasma Techniques on Cancer Attenuation. Cancers (Basel) 2019; 11:E1030. [PMID: 31336648 PMCID: PMC6678366 DOI: 10.3390/cancers11071030] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022] Open
Abstract
For the last few decades, while significant improvements have been achieved in cancer therapy, this family of diseases is still considered one of the deadliest threats to human health. Thus, there is an urgent need to find novel strategies in order to tackle this vital medical issue. One of the most pivotal causes of cancer initiation is the presence of reactive oxygen species (ROS) inside the body. Interestingly, on the other hand, high doses of ROS possess the capability to damage malignant cells. Moreover, several important intracellular mechanisms occur during the production of ROS. For these reasons, inducing ROS inside the biological system by utilizing external physical or chemical methods is a promising approach to inhibit the growth of cancer cells. Beside conventional technologies, cold atmospheric plasmas are now receiving much attention as an emerging therapeutic tool for cancer treatment due to their unique biophysical behavior, including the ability to generate considerable amounts of ROS. This review summarizes the important mechanisms of ROS generated by chemical, physical, and plasma approaches. We also emphasize the biological effects and cancer inhibition capabilities of ROS.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Linh Nhat Nguyen
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Mahmuda Akter
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Gyungsoon Park
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Bio-display, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea.
| |
Collapse
|
15
|
Crestini A, Vona R, Lo Giudice M, Sbriccoli M, Piscopo P, Borrelli A, Rivabene R, Ricceri L, Mancini A, Confaloni A. Differentiation-Dependent Effects of a New Recombinant Manganese Superoxide Dismutase on Human SK-N-BE Neuron-Like Cells. Neurochem Res 2018; 44:400-411. [PMID: 30471001 DOI: 10.1007/s11064-018-2686-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 09/24/2018] [Accepted: 11/16/2018] [Indexed: 11/29/2022]
Abstract
We have recently isolated a new isoform of recombinant manganese superoxide dismutase (rMnSOD) which provides a potent antitumor activity and strongly counteracts the occurrence of oxidative stress and tissue inflammation. This isoform, in addition to the enzymatic action common to all SODs, also shows special functional and structural properties, essentially due to the presence of a first leader peptide that allows the protein to enter easily into cells. Among endogenous antioxidants, SOD constitutes the first line of natural defence against pathological effects induced by an excess of free radicals. Here, we firstly describe the effects of our rMnSOD administration on the proliferant and malignant undifferentiated human neuroblastoma SK-N-BE cell line. Moreover, we also test the effects of rMnSOD in the all trans retinoic-differentiated SK-N-BE neuron-like cells, a quiescent "not malignant" model. While rMnSOD showed an antitumor activity on proliferating cells, a poor sensitivity to rMnSOD overload in retinoid-differentiated neuron-like cells was observed. However, in the latter case, in presence of experimental-induced oxidative stress, overcharge of rMnSOD enhanced the oxidant effects, through an increase of H2O2 due to low activity of both catalase and glutathione peroxidase. In conclusion, our data show that rMnSOD treatment exerts differential effects, which depend upon both cell differentiation and redox balance, addressing attention to the potential use of the recombinant enzyme on differentiated neurons. These facts ultimately pave the way for further preclinical studies aimed at evaluation of rMnSOD effects in models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessio Crestini
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Rosa Vona
- Biomarkers Unit, Center for Gender-Specific Medicine, Italian National Institute of Health, Rome, Italy
| | - Maria Lo Giudice
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Marco Sbriccoli
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Antonella Borrelli
- Molecular Biology and Viral Oncology Unit, Department of Experimental Oncology, Istituto Nazionale Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Roberto Rivabene
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - Laura Ricceri
- Centre for Behavioural Sciences and Mental Health, Italian National Institute of Health, Rome, Italy
| | - Aldo Mancini
- Leadhexa Biotechnologies Inc., San Francisco, CA, USA
| | - Annamaria Confaloni
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy.
| |
Collapse
|
16
|
Pascotini MET, Flores DAE, Kegler MA, Konzen MV, Fornari MAL, Arend MJ, Gabbi MP, Gobo MLA, Bochi DGV, Prado DALC, de Carvalho DLM, Duarte DMM, da Cruz DIBM, Moresco DRN, dos Santos DARS, Royes DLFF, Fighera DMR. Brain-Derived Neurotrophic Factor Levels are Lower in Chronic Stroke Patients: A Relation with Manganese-dependent Superoxide Dismutase ALA16VAL Single Nucleotide Polymorphism through Tumor Necrosis Factor-α and Caspases Pathways. J Stroke Cerebrovasc Dis 2018; 27:3020-3029. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.06.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/03/2018] [Accepted: 06/24/2018] [Indexed: 12/19/2022] Open
|
17
|
Sokolova M, Sahraoui A, Høyem M, Øgaard J, Lien E, Aukrust P, Yndestad A, Ranheim T, Scholz H. NLRP3 inflammasome mediates oxidative stress-induced pancreatic islet dysfunction. Am J Physiol Endocrinol Metab 2018; 315:E912-E923. [PMID: 30016155 DOI: 10.1152/ajpendo.00461.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Inflammasomes are multiprotein inflammatory platforms that induce caspase-1 activation and subsequently interleukin (IL)-1β and IL-18 processing. The NLRP3 inflammasome is activated by different forms of oxidative stress, and, based on the central role of IL-1β in the destruction of pancreatic islets, it could be related to the development of diabetes. We therefore investigated responses in wild-type C57Bl/6 (WT) mice, NLRP3-/- mice, and mice deficient in apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) after exposing islets to short-term hypoxia or alloxan-induced islet damage. NLRP3-deficient islets compared with WT islets had preserved function ex vivo and were protected against hypoxia-induced cell death. Furthermore, NLRP3 and ASC-deficient mice were protected against oxidative stress-induced diabetes caused by repetitive low-dose alloxan administration, and this was associated with reduced β-cell death and reduced macrophage infiltration. This suggests that the beneficial effect of NLRP3 inflammasome deficiency on oxidative stress-mediated β-cell damage could involve reduced macrophage infiltration and activation. To support the role of macrophage activation in alloxan-induced diabetes, we injected WT mice with liposomal clodronate, which causes macrophage depletion before induction of a diabetic phenotype by alloxan treatment, resulting in improved glucose homeostasis in WT mice. We show here that the NLRP3 inflammasome acts as a mediator of hypoxia and oxidative stress in insulin-producing cells, suggesting that inhibition of the NLRP3 inflammasome could have beneficial effects on β-cell preservation.
Collapse
Affiliation(s)
- Marina Sokolova
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
| | - Afaf Sahraoui
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- Institute for Surgical Research and Section for Transplantation Surgery, Oslo University Hospital , Oslo , Norway
| | - Merete Høyem
- Institute for Surgical Research and Section for Transplantation Surgery, Oslo University Hospital , Oslo , Norway
| | - Jonas Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
| | - Egil Lien
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital , Oslo , Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- K.G. Jebsen Inflammation Research Center, University of Oslo , Oslo , Norway
| | - Hanne Scholz
- Institute of Clinical Medicine, University of Oslo , Oslo , Norway
- Institute for Surgical Research and Section for Transplantation Surgery, Oslo University Hospital , Oslo , Norway
| |
Collapse
|
18
|
da Rocha AL, Teixeira GR, Pinto AP, de Morais GP, Oliveira LDC, de Vicente LG, da Silva LECM, Pauli JR, Cintra DE, Ropelle ER, de Moura LP, Mekary RA, de Freitas EC, da Silva ASR. Excessive training induces molecular signs of pathologic cardiac hypertrophy. J Cell Physiol 2018; 233:8850-8861. [PMID: 29797568 DOI: 10.1002/jcp.26799] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/30/2018] [Indexed: 12/27/2022]
Abstract
Chronic exercise induces cardiac remodeling that promotes left ventricular hypertrophy and cardiac functional improvement, which are mediated by the mammalian or the mechanistic target of rapamycin (mTOR) as well as by the androgen and glucocorticoid receptors (GRs). However, pathological conditions (i.e., chronic heart failure, hypertension, and aortic stenosis, etc.) also induce cardiac hypertrophy, but with detrimental function, high levels of proinflammatory cytokines and myostatin, elevated fibrosis, reduced adenosine monophosphate-activated protein kinase (AMPK) activation, and fetal gene reactivation. Furthermore, recent studies have evidenced that excessive training induced an inflammatory status in the serum, muscle, hypothalamus, and liver, suggesting a pathological condition that could also be detrimental to cardiac tissue. Here, we verified the effects of three running overtraining (OT) models on the molecular parameters related to physiological and pathological cardiac hypertrophy. C57BL/6 mice performed three different OT protocols and were evaluated for molecular parameters related to physiological and pathological cardiac hypertrophy, including immunoblotting, reverse transcription polymerase chain reaction, histology, and immunohistochemistry analyses. In summary, the three OT protocols induced left ventricle (LV) hypertrophy with signs of cardiac fibrosis and negative morphological adaptations. These maladaptations were accompanied by reductions in AMPKalpha (Thr172) phosphorylation, androgen receptor, and GR expressions, as well as by an increase in interleukin-6 expression. Specifically, the downhill running-based OT model reduced the content of some proteins related to the mTOR signaling pathway and upregulated the β-isoform of myosin heavy-chain gene expression, presenting signs of LV pathological hypertrophy development.
Collapse
Affiliation(s)
- Alisson L da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Giovana R Teixeira
- Department of Physical Education, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Ana P Pinto
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Gustavo P de Morais
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Luciana da C Oliveira
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Larissa Gaioto de Vicente
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Lilian E C M da Silva
- Department of Ophthalmology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Leandro P de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Rania A Mekary
- Department of Pharmaceutical Business and Administrative Sciences, MCPHS University, Boston, Massachusetts.,Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ellen C de Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.,School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
19
|
Abstract
The evolution of cardiac disease after an acute ischemic event depends on a complex and dynamic network of mechanisms alternating from ischemic damage due to acute coronary occlusion to reperfusion injury due to the adverse effects of coronary revascularization till post-ischemic remodeling. Cardioprotection is a new purpose of the therapeutic interventions in cardiology with the goal to reduce infarct size and thus prevent the progression toward heart failure after an acute ischemic event. In a complex biological system such as the human one, an effective cardioprotective strategy should diachronically target the network of cross-talking pathways underlying the disease progression. Thyroid system is strictly interconnected with heart homeostasis, and recent studies highlighted its role in cardioprotection, in particular through the preservation of mitochondrial function and morphology, the antifibrotic and proangiogenetic effect and also to the potential induction of cell regeneration and growth. The objective of this review was to highlight the cardioprotective role of triiodothyronine in the complexity of post-ischemic disease evolution.
Collapse
|
20
|
Tumor necrosis factor-α receptor 1 contributes to ethanol-induced vascular reactive oxygen species generation and hypertension. ACTA ACUST UNITED AC 2017; 11:684-696.e3. [DOI: 10.1016/j.jash.2017.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/22/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023]
|
21
|
Lee H, Kim EK, Kim HY, Kim TI. Effects of Exposure to Ozone on the Ocular Surface in an Experimental Model of Allergic Conjunctivitis. PLoS One 2017; 12:e0169209. [PMID: 28046113 PMCID: PMC5207719 DOI: 10.1371/journal.pone.0169209] [Citation(s) in RCA: 16] [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: 08/24/2016] [Accepted: 12/13/2016] [Indexed: 12/30/2022] Open
Abstract
Based on previous findings that ozone can induce an inflammatory response in the ocular surface of an animal model and in cultured human conjunctival epithelial cells, we investigated whether exposure to ozone exacerbates symptoms of allergic conjunctivitis. We evaluated the effects of exposure to ozone on conjunctival chemosis, conjunctival injection, corneal and conjunctival fluorescein staining scores, production of inflammatory cytokines in tears, and aqueous tear production in a mouse model of allergic conjunctivitis. To validate our in vivo results, we used interleukin (IL)-1α-pretreated conjunctival epithelial cells as an in vitro substitute for the mouse model. We evaluated whether exposure to ozone increased the inflammatory response and altered oxidative status and mitochondrial function in IL-1α-pretreated conjunctival epithelial cells. In the in vivo study, ozone induced increases in conjunctival chemosis, conjunctival injection, corneal and conjunctival fluorescein staining scores, and production of inflammatory cytokines, accompanied by a decrease in tear volume. In the in vitro study, exposure to ozone led to additional increases in IL-6 and tumor necrosis factor-α mRNA levels, which were already induced by treatment with IL-1α. Ozone did not induce any changes in cell viability. Pretreatment with IL-1α increased the expression of manganese superoxide dismutase, and exposure to ozone led to additional increments in the expression of this antioxidant enzyme. Ozone did not induce any changes in mitochondrial activity or expression of mitochondrial enzymes and proteins related to mitochondrial function, with the exception of phosphor-mammalian target of rapamycin. Treatment with butylated hydroxyanisole, a free radical scavenger, attenuated the ozone-induced increases in IL-6 expression in IL-1α-pretreated conjunctival epithelial cells. Therefore, we conclude that exposure to ozone exacerbates the detrimental effects on the integrity of the ocular surface caused by conjunctival allergic reactions, and further increases the inflammatory response in IL-1α-pretreated conjunctival epithelial cells.
Collapse
Affiliation(s)
- Hun Lee
- Department of Ophthalmology, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, South Korea
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Eung Kweon Kim
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
- Department of Ophthalmology, Corneal Dystrophy Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Hee Young Kim
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
- Department of Ophthalmology, Corneal Dystrophy Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae-im Kim
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
- Department of Ophthalmology, Corneal Dystrophy Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- * E-mail:
| |
Collapse
|
22
|
Ongali B, Nicolakakis N, Tong XK, Aboulkassim T, Imboden H, Hamel E. Enalapril Alone or Co-Administered with Losartan Rescues Cerebrovascular Dysfunction, but not Mnemonic Deficits or Amyloidosis in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2016; 51:1183-95. [PMID: 26923013 DOI: 10.3233/jad-150868] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The co-administration of angiotensin converting enzyme inhibitors (ACEi) and angiotensin II (AngII) receptor blockers (ARB) that bind angiotensin type 1 receptors (AT1R) may protect from Alzheimer's disease (AD) better than each treatment taken alone. We tested the curative potential of the non brain-penetrant ACEi enalapril (3 mg/kg/day) administered for 3 months either alone or in combination with the brain penetrant ARB losartan (10 mg/kg/day) in aged (∼15 months) transgenic mice overexpressing a mutated form of the human amyloid-β protein precursor (AβPP, thereafter APP mice). We studied cerebrovascular function, protein levels of oxidative stress markers (superoxide dismutases SOD1, SOD2 and the NADPH oxidase subunit p67phox), amyloid-β (Aβ) pathology, astrogliosis, cholinergic innervation, AT1R and angiotensin IV receptor (AT4R) levels, together with cognitive performance. Both treatments normalized cerebrovascular reactivity and p67phox protein levels, but they did not reduce the cerebrovascular levels of SOD1. Combined treatment normalized cerebrovascular SOD2 levels, significantly attenuated astrogliosis, but did not reduce the increased levels of cerebrovascular AT1R. Yet, combined therapy enhanced thioflavin-S labeled Aβ plaque burden, a tendency not significant when Aβ1 - 42 plaque load was considered. None of the treatments rescued cognitive deficits, cortical AT4R or cholinergic innervation. We conclude that both treatments normalized cerebrovascular function by inhibiting the AngII-induced oxidative stress cascade, and that the positive effects of the combined therapy on astrogliosis were likely due to the ability of losartan to enter brain parenchyma. However, enalapril did not potentiate, and may even dampen, the reported cognitive benefits of losartan, raising caution when selecting the most appropriate antihypertensive therapy in AD patients.
Collapse
Affiliation(s)
- Brice Ongali
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Nektaria Nicolakakis
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Xing-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Tahar Aboulkassim
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Hans Imboden
- Institute of Cell Biology, University of Bern, Switzerland
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
23
|
Alam AHMK, Hossain ASMS, Khan MA, Kabir SR, Reza MA, Rahman MM, Islam MS, Rahman MAA, Rashid M, Sadik MG. The Antioxidative Fraction of White Mulberry Induces Apoptosis through Regulation of p53 and NFκB in EAC Cells. PLoS One 2016; 11:e0167536. [PMID: 27936037 PMCID: PMC5147903 DOI: 10.1371/journal.pone.0167536] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
In this study, the antioxidative fraction of white mulberry (Morus alba) was found to have an apotogenic effect on Ehrlich's ascites carcinoma cell-induced mice (EAC mice) that correlate with upregulated p53 and downregulated NFκB signaling. The antioxidant activities and polyphenolic contents of various mulberry fractions were evaluated by spectrophotometry and the ethyl acetate fraction (EAF) was selected for further analysis. Strikingly, the EAF caused 70.20% tumor growth inhibition with S-phase cell cycle arrest, normalized blood parameters including red/white blood cell counts and suppressed the tumor weight of EAC mice compared with untreated controls. Fluorescence microscopy analysis of EAF-treated EAC cells revealed DNA fragmentation, cell shrinkage, and plasma membrane blebbing. These characteristic morphological features of apoptosis influenced us to further investigate pro- and anti-apoptotic signals in EAF-treated EAC mice. Interestingly, apoptosis correlated with the upregulation of p53 and its target genes PARP-1 and Bax, and also with the down-regulation of NFκB and its target genes Bcl-2 and Bcl-xL. Our results suggest that the tumor- suppressive effect of the antioxidative fraction of white mulberry is likely due to apoptosis mediated by p53 and NFκB signaling.
Collapse
Affiliation(s)
- AHM Khurshid Alam
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
- * E-mail:
| | | | - Muhammad Ali Khan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Syed Rashel Kabir
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Md Abu Reza
- Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | | | | | | | - Mamunur Rashid
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Md Golam Sadik
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
24
|
Pereira EJ, Smolko CM, Janes KA. Computational Models of Reactive Oxygen Species as Metabolic Byproducts and Signal-Transduction Modulators. Front Pharmacol 2016; 7:457. [PMID: 27965578 PMCID: PMC5126069 DOI: 10.3389/fphar.2016.00457] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/14/2016] [Indexed: 12/30/2022] Open
Abstract
Reactive oxygen species (ROS) are widely involved in intracellular signaling and human pathologies, but their precise roles have been difficult to enumerate and integrate holistically. The context- and dose-dependent intracellular effects of ROS can lead to contradictory experimental results and confounded interpretations. For example, lower levels of ROS promote cell signaling and proliferation, whereas abundant ROS cause overwhelming damage to biomolecules and cellular apoptosis or senescence. These complexities raise the question of whether the many facets of ROS biology can be joined under a common mechanistic framework using computational modeling. Here, we take inventory of some current models for ROS production or ROS regulation of signaling pathways. Several models captured non-intuitive observations or made predictions that were later verified by experiment. There remains a need for systems-level analyses that jointly incorporate ROS production, handling, and modulation of multiple signal-transduction cascades.
Collapse
Affiliation(s)
- Elizabeth J Pereira
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, USA
| | - Christian M Smolko
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, USA
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville VA, USA
| |
Collapse
|
25
|
Jabbar A, Pingitore A, Pearce SHS, Zaman A, Iervasi G, Razvi S. Thyroid hormones and cardiovascular disease. Nat Rev Cardiol 2016; 14:39-55. [PMID: 27811932 DOI: 10.1038/nrcardio.2016.174] [Citation(s) in RCA: 438] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Myocardial and vascular endothelial tissues have receptors for thyroid hormones and are sensitive to changes in the concentrations of circulating thyroid hormones. The importance of thyroid hormones in maintaining cardiovascular homeostasis can be deduced from clinical and experimental data showing that even subtle changes in thyroid hormone concentrations - such as those observed in subclinical hypothyroidism or hyperthyroidism, and low triiodothyronine syndrome - adversely influence the cardiovascular system. Some potential mechanisms linking the two conditions are dyslipidaemia, endothelial dysfunction, blood pressure changes, and direct effects of thyroid hormones on the myocardium. Several interventional trials showed that treatment of subclinical thyroid diseases improves cardiovascular risk factors, which implies potential benefits for reducing cardiovascular events. Over the past 2 decades, accumulating evidence supports the association between abnormal thyroid function at the time of an acute myocardial infarction (MI) and subsequent adverse cardiovascular outcomes. Furthermore, experimental studies showed that thyroid hormones can have an important therapeutic role in reducing infarct size and improving myocardial function after acute MI. In this Review, we summarize the literature on thyroid function in cardiovascular diseases, both as a risk factor as well as in the setting of cardiovascular diseases such as heart failure or acute MI, and outline the effect of thyroid hormone replacement therapy for reducing the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Avais Jabbar
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.,Freeman Hospital, Freeman Rd, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | | | - Simon H S Pearce
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.,Department of Endocrinology, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK
| | - Azfar Zaman
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.,Freeman Hospital, Freeman Rd, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | - Giorgio Iervasi
- Clinical Physiology Institute, CNR, Via Moruzzi 1, 56124, Pisa, Italy
| | - Salman Razvi
- Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.,Gateshead Health NHS Foundation Trust, Saltwell Road South, Gateshead NE8 4YL, UK
| |
Collapse
|
26
|
Trippel TD, Holzendorf V, Halle M, Gelbrich G, Nolte K, Duvinage A, Schwarz S, Rutscher T, Wiora J, Wachter R, Herrmann-Lingen C, Duengen HD, Hasenfuß G, Pieske B, Edelmann F. Ghrelin and hormonal markers under exercise training in patients with heart failure with preserved ejection fraction: results from the Ex-DHF pilot study. ESC Heart Fail 2016; 4:56-65. [PMID: 28217313 PMCID: PMC5292632 DOI: 10.1002/ehf2.12109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 07/06/2016] [Accepted: 07/19/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Over 50% of patients with symptomatic heart failure (HF) experience HF with preserved ejection fraction (HFpEF). Exercise training (ET) is effective in improving cardiorespiratory fitness and dimensions of quality of life in patients with HFpEF. A systemic pro-inflammatory state induced by comorbidities as the cause of myocardial structural and functional alterations has been proposed in HFpEF. ET modifies myocardial structure and has been related to inflammatory state. We investigated Ghrelin, related adipokines, markers of inflammation, and neuro-hormonal activation in patients undergoing a structured ET vs. usual care are with HFpEF. METHODS AND RESULTS Ex-DHF-P was a prospective, controlled, randomized multi-centre trial on structured and supervised ET in patients with HFpEF. We performed a post hoc analysis in 62 patients from Ex-DHF-P. Ghrelin, adiponectin, leptin, IL-1ß, IL-6, IL-10, tumour necrosis factor-alpha, MR-proANP, MR-proADM, CT-proET1, and CT-proAVP were assessed to seize the impact of ET on these markers in patients with HFpEF. Thirty-six (58%) patients were female, mean age was 64 years, and median ghrelin was 928 pg/mL (interquartile range 755-1156). When stratified for high versus low ghrelin, groups significantly differed at baseline in presence obesity, waist circumference, and adiponectin levels (P < 0.05, respectively). Overall, ghrelin levels rose significantly to 1013 pg/mL (interquartile range 813-1182) (P < 0.001). Analysis of covariance modelling for change in ghrelin identified ET (P = 0.013) and higher baseline adiponectin levels (P = 0.035) as influencing factors. CONCLUSIONS Exercise training tended to increase ghrelin levels in Ex-DHF-P. This increase was especially pronounced in patients with higher baseline adiponectin levels. Future trials are needed to investigate the effect of ET on endogenous ghrelin levels in regard to interactions with cardiac structure and clinically meaningful surrogate parameters.
Collapse
Affiliation(s)
- Tobias Daniel Trippel
- Department of Internal Medicine - CardiologyCharité - Universitaetsmedizin Berlin, Campus Virchow KlinikumBerlinGermany; DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
| | - Volker Holzendorf
- ZKS Leipzig (Clinical Trial Centre Leipzig) University of Leipzig Leipzig Germany
| | - Martin Halle
- DZHK (German Centre for Cardiovascular Research), Partner Site MunichMunich Heart AllianceMunichGermany; Else Kröner-Fresenius-Zentrum, Klinikum rechts der IsarTechnical University MunichMunichGermany
| | - Götz Gelbrich
- Institute for Clinical Epidemiology and BiometryUniversity of WürzburgWürzburgGermany; Clinical Trial CentreUniversity Hospital WürzburgWürzburgGermany
| | - Kathleen Nolte
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CentreGöttingenGermany; DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Andre Duvinage
- DZHK (German Centre for Cardiovascular Research), Partner Site MunichMunich Heart AllianceMunichGermany; Else Kröner-Fresenius-Zentrum, Klinikum rechts der IsarTechnical University MunichMunichGermany; Department of Cardiology and PneumologyUniversity of Göttingen Medical CentreGöttingenGermany
| | - Silja Schwarz
- DZHK (German Centre for Cardiovascular Research), Partner Site MunichMunich Heart AllianceMunichGermany; Else Kröner-Fresenius-Zentrum, Klinikum rechts der IsarTechnical University MunichMunichGermany
| | - Tinka Rutscher
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CentreGöttingenGermany; DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Julian Wiora
- Department of Internal Medicine - Cardiology Charité - Universitaetsmedizin Berlin, Campus Virchow Klinikum Berlin Germany
| | - Rolf Wachter
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CentreGöttingenGermany; DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Christoph Herrmann-Lingen
- DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany; Department for Psychosomatic Medicine and PsychotherapyUniversity of Göttingen Medical CentreGöttingenGermany
| | - Hans-Dirk Duengen
- Department of Internal Medicine - CardiologyCharité - Universitaetsmedizin Berlin, Campus Virchow KlinikumBerlinGermany; DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
| | - Gerd Hasenfuß
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CentreGöttingenGermany; DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Burkert Pieske
- Department of Internal Medicine - CardiologyCharité - Universitaetsmedizin Berlin, Campus Virchow KlinikumBerlinGermany; DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
| | - Frank Edelmann
- Department of Internal Medicine - CardiologyCharité - Universitaetsmedizin Berlin, Campus Virchow KlinikumBerlinGermany; DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany; Department of Cardiology and PneumologyUniversity of Göttingen Medical CentreGöttingenGermany
| |
Collapse
|
27
|
Kelly FJ, Postle AD, Phillips GJ. Neutrophils and oxygen-induced lung injury: a case of when a few is still too many. Redox Rep 2016; 1:37-44. [DOI: 10.1080/13510002.1994.11746954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
28
|
Maternal Obesity Promotes Diabetic Nephropathy in Rodent Offspring. Sci Rep 2016; 6:27769. [PMID: 27277011 PMCID: PMC4899795 DOI: 10.1038/srep27769] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 05/25/2016] [Indexed: 02/07/2023] Open
Abstract
Maternal obesity is known to increase the risk of obesity and diabetes in offspring. Though diabetes is a key risk factor for the development of chronic kidney disease (CKD), the relationship between maternal obesity and CKD has not been clearly defined. In this study, a mouse model of maternal obesity was employed to determine the impact of maternal obesity on development of diabetic nephropathy in offspring. Female C57BL/6 mice were fed high-fat diet (HFD) for six weeks prior to mating, during gestation and lactation. Male offspring were weaned to normal chow diet. At postnatal Week 8, offspring were randomly administered low dose streptozotocin (STZ, 55 mg/kg/day for five days) to induce diabetes. Assessment of renal damage took place at postnatal Week 32. We found that offspring of obese mothers had increased renal fibrosis, inflammation and oxidative stress. Importantly, offspring exposed to maternal obesity had increased susceptibility to renal damage when an additional insult, such as STZ-induced diabetes, was imposed. Specifically, renal inflammation and oxidative stress induced by diabetes was augmented by maternal obesity. Our findings suggest that developmental programming induced by maternal obesity has implications for renal health in offspring. Maternal obesity should be considered a risk factor for CKD.
Collapse
|
29
|
Issop L, Ostuni MA, Lee S, Laforge M, Péranzi G, Rustin P, Benoist JF, Estaquier J, Papadopoulos V, Lacapère JJ. Translocator Protein-Mediated Stabilization of Mitochondrial Architecture during Inflammation Stress in Colonic Cells. PLoS One 2016; 11:e0152919. [PMID: 27054921 PMCID: PMC4824355 DOI: 10.1371/journal.pone.0152919] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/21/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Chronic inflammation of the gastrointestinal tract increasing the risk of cancer has been described to be linked to the high expression of the mitochondrial translocator protein (18 kDa; TSPO). Accordingly, TSPO drug ligands have been shown to regulate cytokine production and to improve tissue reconstruction. We used HT-29 human colon carcinoma cells to evaluate the role of TSPO and its drug ligands in tumor necrosis factor (TNF)-induced inflammation. TNF-induced interleukin (IL)-8 expression, coupled to reactive oxygen species (ROS) production, was followed by TSPO overexpression. TNF also destabilized mitochondrial ultrastructure, inducing cell death by apoptosis. Treatment with the TSPO drug ligand PK 11195 maintained the mitochondrial ultrastructure, reducing IL-8 and ROS production and cell death. TSPO silencing and overexpression studies demonstrated that the presence of TSPO is essential to control IL-8 and ROS production, so as to maintain mitochondrial ultrastructure and to prevent cell death. Taken together, our data indicate that inflammation results in the disruption of mitochondrial complexes containing TSPO, leading to cell death and epithelia disruption. SIGNIFICANCE This work implicates TSPO in the maintenance of mitochondrial membrane integrity and in the control of mitochondrial ROS production, ultimately favoring tissue regeneration.
Collapse
Affiliation(s)
- Leeyah Issop
- Sorbonne Universités – Université Pierre et Marie Curie Université de Paris VI, École Normale Supérieure – PSL Research University, Département de Chimie, CNRS UMR 7203 LBM, 4 Place Jussieu, F-75005, Paris, France
- The Research Institute of the McGill University Health Center and the Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Mariano A. Ostuni
- INSERM UMRS 1134, Institut National de la Transfusion Sanguine, 6 rue Alexandre Cabanel, Université Paris 7 Denis Diderot, F-75015 Paris, France
| | - Sunghoon Lee
- The Research Institute of the McGill University Health Center and the Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | | | - Gabriel Péranzi
- Sorbonne Universités – Université Pierre et Marie Curie Université de Paris VI, École Normale Supérieure – PSL Research University, Département de Chimie, CNRS UMR 7203 LBM, 4 Place Jussieu, F-75005, Paris, France
| | - Pierre Rustin
- INSERM UMR 1141, Hôpital Robert Debré, and Université Paris 7 Denis Diderot, F-75019, Paris, France
| | - Jean-François Benoist
- INSERM UMR 1141, Hôpital Robert Debré, and Université Paris 7 Denis Diderot, F-75019, Paris, France
| | - Jérome Estaquier
- CNRS FR 3636, Université Paris Descartes, Paris, France
- Université Laval, Faculté de Médecine, Département de microbiologie-infectiologie et d’immunologie, Quebec City, Quebec, G1V06A, Canada
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Center and the Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Jean-Jacques Lacapère
- Sorbonne Universités – Université Pierre et Marie Curie Université de Paris VI, École Normale Supérieure – PSL Research University, Département de Chimie, CNRS UMR 7203 LBM, 4 Place Jussieu, F-75005, Paris, France
- * E-mail:
| |
Collapse
|
30
|
Pingitore A, Iervasi G, Forini F. Role of the Thyroid System in the Dynamic Complex Network of Cardioprotection. Eur Cardiol 2016; 11:36-42. [PMID: 30310446 DOI: 10.15420/ecr.2016:9:2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cardioprotection is a common goal of new therapeutic strategies in patients with coronary artery disease and/or left ventricular dysfunction. Myocardial damage following ischaemia/reperfusion injury lead to left ventricular adverse remodelling through many mechanisms arising from different cell types in different myocardial districts, namely the border and remote zone. Cardioprotection must face this complex, dynamic network of cooperating units. In this scenario, thyroid hormones can represent an effective therapeutic strategy due to the numerous actions and regulating mechanisms carried out at the level of the myocytes, interstitium and the vasculature, as well as to the activation of different pro-survival intracellular pathways involved in cardioprotection.
Collapse
Affiliation(s)
| | - Giorgio Iervasi
- Clinical Physiology Institute, National Research Council (CNR), Pisa, Italy
| | - Francesca Forini
- Clinical Physiology Institute, National Research Council (CNR), Pisa, Italy
| |
Collapse
|
31
|
Ghandadi M, Behravan J, Abnous K, Mosaffa F. Reactive Oxygen Species Mediate TNF-⍺ Cytotoxic Effects in the Multidrug-Resistant Breast Cancer Cell Line MCF-7/MX. Oncol Res Treat 2015; 39:54-9. [DOI: 10.1159/000442144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/22/2015] [Indexed: 11/19/2022]
|
32
|
Koike A, Minamiguchi I, Fujimori K, Amano F. Nitric oxide is an important regulator of heme oxygenase-1 expression in the lipopolysaccharide and interferon-γ-treated murine macrophage-like cell line J774.1/JA-4. Biol Pharm Bull 2015; 38:7-16. [PMID: 25744452 DOI: 10.1248/bpb.b14-00405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. The HO-1 products provide antioxidant cytoprotection in addition to having potent antiinflammatory and immunomodulatory functions. HO-1 is induced by its substrate heme and environmental factors including oxidative and heat stresses. Although previous studies reported that lipopolysaccharide (LPS) induced the expression of both the HO-1 gene and its protein in macrophages, the major regulators of HO-1 expression remain unknown. To identify these regulators, we used two types of cell, the murine macrophage-like cell line J774.1/JA-4 and its LPS-resistant mutant, LPS1916. Based on a comparison of the results obtained with these cells, we found that nitric oxide (NO) was closely linked to the induction of HO-1. Real-time polymerase chain reaction (PCR) showed that the time course for inducible HO-1 mRNA by LPS or LPS+interferon (IFN)-γ was similar to that for inducible NO synthase (iNOS) mRNA. Furthermore, the expression of iNOS mRNA and protein increased earlier than that of HO-1 mRNA and protein. N-Nitro-L-arginine methyl ester, an NO synthase inhibitor, reduced both HO-1 expression and NO production in LPS+IFN-γ-treated JA-4 cells. Furthermore, NOC-12, an NO donor, significantly induced HO-1 expression not only in JA-4 but also in LPS1916 cells. Reactive oxygen species (ROS) scavengers, such as superoxide dismutase and catalase, did not affect HO-1 protein expression in LPS+IFN-γ-treated JA-4 cells. These results suggest that, among ROS, NO plays an important role in HO-1 induction in activated macrophages treated with LPS+IFN-γ.
Collapse
Affiliation(s)
- Atsushi Koike
- Laboratory of Biodefense & Regulation, Osaka University of Pharmaceutical Sciences
| | | | | | | |
Collapse
|
33
|
Kundumani-Sridharan V, Subramani J, Das KC. Thioredoxin Activates MKK4-NFκB Pathway in a Redox-dependent Manner to Control Manganese Superoxide Dismutase Gene Expression in Endothelial Cells. J Biol Chem 2015; 290:17505-19. [PMID: 26028649 DOI: 10.1074/jbc.m115.660365] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Indexed: 11/06/2022] Open
Abstract
The mitogen-activated protein kinase kinase 4 (MKK4) is activated via phosphorylation of Ser-257 and Thr-261 by upstream MAP3Ks and activates JNK and p38 MAPKs in response to cellular stress. We show that thioredoxin (Trx), a cellular redox protein, activates MKK4 via Cys-246 and Cys-266 residues as mutation of these residues renders MKK4 insensitive to phosphorylation by MAP3Ks, TNFα, or Trx. MKK4 is activated in vitro by reduced Trx but not oxidized Trx in the absence of an upstream kinase, suggesting that autophosphorylation of this protein occurs due to reduction of Cys-246 and Cys-266 by Trx. Additionally, mutation of Cys-246 and Cys-266 resulted in loss of kinase activity suggesting that the redox state of Cys-246 and Cys-266 is a critical determinant of MKK4 activation. Trx induces manganese superoxide dismutase (MnSOD) gene transcription by activating MKK4 via redox control of Cys-246 and Cys-266, as mutation of these residues abrogates MKK4 activation and MnSOD expression. We further show that MKK4 activates NFκB for its binding to the MnSOD promoter, which leads to AP-1 dissociation followed by MnSOD transcription. Taken together, our studies show that the redox status of Cys-246 and Cys-266 in MKK4 controls its activities independent of MAP3K, demonstrating integration of the endothelial redox environment to MAPK signaling.
Collapse
Affiliation(s)
- Venkatesh Kundumani-Sridharan
- From the Department of Anesthesiology and Center for Excellence in Cardiovascular Research, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Jaganathan Subramani
- From the Department of Anesthesiology and Center for Excellence in Cardiovascular Research, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Kumuda C Das
- From the Department of Anesthesiology and Center for Excellence in Cardiovascular Research, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| |
Collapse
|
34
|
Mao Y, Tokudome T, Kishimoto I, Otani K, Nishimura H, Yamaguchi O, Otsu K, Miyazato M, Kangawa K. Endogenous ghrelin attenuates pressure overload-induced cardiac hypertrophy via a cholinergic anti-inflammatory pathway. Hypertension 2015; 65:1238-44. [PMID: 25870195 DOI: 10.1161/hypertensionaha.114.04864] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/01/2015] [Indexed: 01/20/2023]
Abstract
Cardiac hypertrophy, which is commonly caused by hypertension, is a major risk factor for heart failure and sudden death. Endogenous ghrelin has been shown to exert a beneficial effect on cardiac dysfunction and postinfarction remodeling via modulation of the autonomic nervous system. However, ghrelin's ability to attenuate cardiac hypertrophy and its potential mechanism of action are unknown. In this study, cardiac hypertrophy was induced by transverse aortic constriction in ghrelin knockout mice and their wild-type littermates. After 12 weeks, the ghrelin knockout mice showed significantly increased cardiac hypertrophy compared with wild-type mice, as evidenced by their significantly greater heart weight/tibial length ratios (9.2±1.9 versus 7.9±0.8 mg/mm), left ventricular anterior wall thickness (1.3±0.2 versus 1.0±0.2 mm), and posterior wall thickness (1.1±0.3 versus 0.9±0.1 mm). Furthermore, compared with wild-type mice, ghrelin knockout mice showed suppression of the cholinergic anti-inflammatory pathway, as indicated by reduced parasympathetic nerve activity and higher plasma interleukin-1β and interleukin-6 levels. The administration of either nicotine or ghrelin activated the cholinergic anti-inflammatory pathway and attenuated cardiac hypertrophy in ghrelin knockout mice. In conclusion, our results show that endogenous ghrelin plays a crucial role in the progression of pressure overload-induced cardiac hypertrophy via a mechanism that involves the activation of the cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Yuanjie Mao
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Takeshi Tokudome
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Ichiro Kishimoto
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.).
| | - Kentaro Otani
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Hirohito Nishimura
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Osamu Yamaguchi
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Kinya Otsu
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Mikiya Miyazato
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| | - Kenji Kangawa
- From the Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan (Y.M., T.T., I.K., H.N., M.M., K.K.); Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute (K.O.), Department of Cardiovascular Medicine, Graduate School of Medicine (O.Y.), Osaka University, Suita, Japan; and Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (K.O.)
| |
Collapse
|
35
|
Guo T, Zhang L, Konermann A, Zhou H, Jin F, Liu W. Manganese superoxide dismutase is required to maintain osteoclast differentiation and function under static force. Sci Rep 2015; 5:8016. [PMID: 25619900 PMCID: PMC4306132 DOI: 10.1038/srep08016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/24/2014] [Indexed: 02/06/2023] Open
Abstract
Bone homeostasis is maintained by the balance of osteoblasts (OBs) and osteoclasts (OCs). Increased activity of OCs not only contributes to pathological bone resorption, such as osteoporosis and periodontitis, but also is responsible for physiological conditions like orthodontic tooth movement (OTM). However, the detailed mechanism by which orthodontic force promotes the formation of OCs is still poorly understood. In this study, we confirmed that static force promoted the differentiation of human cord monocytes (HMNCs) into OCs depending on loading time and magnitude. Protein expression profiles among HMNCs, HMNCs subjected to static force and mature OCs were established via 2-DE and MALDI-TOF-MS analyses. Total respective protein spot numbers of 549 ± 13, 612 ± 19 and 634 ± 16 were detected in each of the gels by image analysis. The five proteins identified were plasminogen activator inhibitor 2 (PAI-2, Spot 1), peroxiredoxin-6 (PRD-6, Spot 3), manganese superoxide dismutase (SOD2, Spot 6), Rho GDP-dissociation inhibitor 2 (Rho-GDI2, Spot 11) and L-lactate dehydrogenase B chain (L-LDH, Spot 15). More importantly, we revealed that SOD2 was required to maintain monocyte differentiation into functional OCs and may become a potential target for regulating the efficiency of OTM in the future.
Collapse
Affiliation(s)
- Tao Guo
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Liqiang Zhang
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Anna Konermann
- Department of Orthodontics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Hong Zhou
- Department of Orthodontics, Hospital of Stomatology, Xi'an jiaotong University
| | - Fang Jin
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Wenjia Liu
- State Key Laboratory of Military Stomatology, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| |
Collapse
|
36
|
Dave KA, Norris EL, Bukreyev AA, Headlam MJ, Buchholz UJ, Singh T, Collins PL, Gorman JJ. A comprehensive proteomic view of responses of A549 type II alveolar epithelial cells to human respiratory syncytial virus infection. Mol Cell Proteomics 2014; 13:3250-69. [PMID: 25106423 PMCID: PMC4256481 DOI: 10.1074/mcp.m114.041129] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/16/2014] [Indexed: 11/06/2022] Open
Abstract
Human respiratory syncytial virus is a major respiratory pathogen for which there are no suitable antivirals or vaccines. A better understanding of the host cell response to this virus may redress this problem. The present report concerns analysis of multiple independent biological replicates of control and 24 h infected lysates of A549 cells by two different proteomic workflows. One workflow involved fractionation of lysates by in-solution protein IEF and individual fractions were digested using trypsin prior to capillary HPLC-LTQ-OrbitrapXL-MS/MS. A second workflow involved digestion of whole cell lysates and analysis by nanoUltraHPLC-LTQ-OrbitrapElite-MS/MS. Both workflows resulted in the quantification of viral proteins exclusively in lysates of infected cells in the relative abundances anticipated from previous studies. Unprecedented numbers (3247 - 5010) of host cell protein groups were also quantified and the infection-specific regulation of a large number (191) of these protein groups was evident based on a stringent false discovery rate cut-off (<1%). Bioinformatic analyses revealed that most of the regulated proteins were potentially regulated by type I, II, and III interferon, TNF-α and noncanonical NF-κB2 mediated antiviral response pathways. Regulation of specific protein groups by infection was validated by quantitative Western blotting and the cytokine-/key regulator-specific nature of their regulation was confirmed by comparable analyses of cytokine treated A549 cells. Overall, it is evident that the workflows described herein have produced the most comprehensive proteomic characterization of host cell responses to human respiratory syncytial virus published to date. These workflows will form the basis for analysis of the impacts of specific genes of human respiratory syncytial virus responses of A549 and other cell lines using a gene-deleted version of the virus. They should also prove valuable for the analysis of the impact of other infectious agents on host cells.
Collapse
Affiliation(s)
- Keyur A Dave
- From the ‡Protein Discovery Centre, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4029 Australia and
| | - Emma L Norris
- From the ‡Protein Discovery Centre, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4029 Australia and
| | - Alexander A Bukreyev
- §Respiratory Virus Section, Laboratory of Infectious Diseases, National Institute for Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892
| | - Madeleine J Headlam
- From the ‡Protein Discovery Centre, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4029 Australia and
| | - Ursula J Buchholz
- §Respiratory Virus Section, Laboratory of Infectious Diseases, National Institute for Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892
| | - Toshna Singh
- From the ‡Protein Discovery Centre, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4029 Australia and
| | - Peter L Collins
- §Respiratory Virus Section, Laboratory of Infectious Diseases, National Institute for Allergy and Infectious Diseases, NIH, Bethesda, Maryland 20892
| | - Jeffrey J Gorman
- From the ‡Protein Discovery Centre, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4029 Australia and
| |
Collapse
|
37
|
Abstract
The extracellular matrix (ECM) is a living network of proteins that maintains the structural integrity of the myocardium and allows the transmission of electrical and mechanical forces between the myocytes for systole and diastole. During ventricular remodeling, as a result of iterations in the hemodynamic workload, collagen, the main component of the ECM, increases and occupies the areas between the myocytes and the vessels. The resultant fibrosis (reparative fibrosis) is initially a compensatory mechanism and may progress adversely influencing tissue stiffness and ventricular function. Replacement fibrosis appears at sites of previous cardiomyocyte necrosis to preserve the structural integrity of the myocardium, but with the subsequent formation of scar tissue and widespread distribution, it has adverse functional consequences. Continued accumulation of collagen impairs diastolic function and compromises systolic mechanics. Nevertheless, the development of fibrosis is a dynamic process wherein myofibroblasts, the principal cellular elements of fibrosis, are not only metabolically active and capable of the production and upregulation of cytokines but also have contractile properties. During the process of reverse remodeling with left ventricular assist device unloading, cellular, structural, and functional improvements are observed in terminal heart failure patients. With the advent of anti-fibrotic pharmacologic therapies, cellular therapy, and ventricular support devices, fibrosis has become an important therapeutic target in heart failure patients. Herein, we review the current concepts of fibrosis as a main component of ventricular remodeling in heart failure patients. Our aim is to integrate the histopathologic process of fibrosis with the neurohormonal, cytochemical, and molecular changes that lead to ventricular remodeling and its physiologic consequences in patients. The concept of fibrosis as living scar allows us to envision targeting this scar as a means of improving ventricular function in heart failure patients.
Collapse
Affiliation(s)
- Ana Maria Segura
- Department of Cardiovascular Pathology Research, Texas Heart Institute at St. Luke's Episcopal Hospital, MC 1-283, PO Box 20345, Houston, TX, 77225-0345, USA,
| | | | | |
Collapse
|
38
|
Sabatino L, Iervasi G, Pingitore A. Thyroid hormone and heart failure: from myocardial protection to systemic regulation. Expert Rev Cardiovasc Ther 2014; 12:1227-36. [PMID: 25220579 DOI: 10.1586/14779072.2014.957674] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Heart failure (HF) is an intriguing model of chronic disease. It starts as an organ disorder developing, in its progression, into a systemic disease in which the dysfunction of other organs plays a relevant clinical and prognostic impact. Furthermore, continuous activation of systemic pathways plays a role in disease progression, switching their effect from protective to harmful. In this combination of organ dysfunction and systemic derangement, thyroid hormone (TH) have an important regulative impact from cardiovascular to systemic level and from molecular/cellular processes to clinical setting. Whether it is accepted to include TH and thyroid stimulating hormone assessment in the clinical HF course, the next challenge will be to ascertain the benefit of TH replacement therapy in HF patients, taking into consideration the type of hormone to administer, dosage and treatment schedule.
Collapse
Affiliation(s)
- Laura Sabatino
- Clinical Physiology Institute, CNR, Via Moruzzi 1, 56124, Pisa, Italy
| | | | | |
Collapse
|
39
|
Xu W, Yang F, Shen X, Fan S, Liu Q, Wang D. Polysaccharide isolated from Parmelia tinctorum ameliorates ionizing irradiation-induced damage in mice. JOURNAL OF RADIATION RESEARCH 2014; 55:641-647. [PMID: 24722682 PMCID: PMC4099985 DOI: 10.1093/jrr/rrt224] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 12/19/2013] [Accepted: 12/20/2013] [Indexed: 06/03/2023]
Abstract
In this study, WPT-A, a type of water-soluble homogeneous lichen polysaccharide, was isolated and purified from Parmelia tinctorum. We investigated whether WPT-A has radioprotective effects when administered before total-body irradiation (TBI). Mice were treated with WPT-A via intraperitoneal injection (i.p.) once per day for three consecutive days prior to 7, 7.5, 8.5, 10 or 10.5-Gy TBI. Our results indicated that the survival rate was enhanced at a range of levels of TBI. The calculated dose reduction factor (DRF) was 1.2. White blood cell (WBC) counts, spleen colony forming units (CFU-S) and bone marrow nucleated cell (BMNC) counts were used to investigate the radioprotective effects of WPT-A on the hematopoietic system. The treatment groups received WPT-A at 20, 50 and 80 mg/kg b.w. doses before 6.5-Gy TBI and showed significantly higher BMNC and WBC counts compared with the radiation-only group. The groups administered 50 and 80 mg/kg b.w. WPT-A showed a significant increase in CFU-S compared with the radiation-only group. We also carried out a single cell gel electrophoresis assay to explore the radioprotective effects of WPT-A on DNA damage. The results from single-cell gel electrophoresis of peripheral blood leukocytes showed that WPT-A attenuated radiation-induced DNA damage. These results indicate a potential use for WPT-A as a radioprotector.
Collapse
Affiliation(s)
- Wenqing Xu
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Fujun Yang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Xiu Shen
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Saijun Fan
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Qiang Liu
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Dezhi Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
40
|
Salisbury TB, Tomblin JK, Primerano DA, Boskovic G, Fan J, Mehmi I, Fletcher J, Santanam N, Hurn E, Morris GZ, Denvir J. Endogenous aryl hydrocarbon receptor promotes basal and inducible expression of tumor necrosis factor target genes in MCF-7 cancer cells. Biochem Pharmacol 2014; 91:390-9. [PMID: 24971714 DOI: 10.1016/j.bcp.2014.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 01/11/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that upon activation by the toxicant 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) stimulates gene expression and toxicity. AHR is also important for normal mouse physiology and may play a role in cancer progression in the absence of environmental toxicants. The objective of this report was to identify AHR-dependent genes (ADGs) whose expression is regulated by AHR in the absence of toxicants. RNA-Seq analysis revealed that AHR regulated the expression of over 600 genes at an FDR<10% in MCF-7 breast cancer cells upon knockdown with short interfering RNA. Pathway analysis revealed that a significant number of ADGs were components of TCDD and tumor necrosis factor (TNF) pathways. We also demonstrated that siRNA knockdown of AHR modulated TNF induction of MNSOD and cytotoxicity in MCF-7 cells. Collectively, the major new findings of this report are: (1) endogenous AHR promotes the expression of xenobiotic metabolizing enzymes even in the absence of toxicants and drugs, (2) AHR by modulating the basal expression of a large fraction of TNF target genes may prime them for TNF stimulation and (3) AHR is required for TNF induction of MNSOD and the cellular response to cytotoxicity in MCF-7 cells. This latter result provides a potentially new role for AHR in MCF-7 cancer progression as a mediator of TNF and antioxidant responses.
Collapse
Affiliation(s)
- Travis B Salisbury
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Justin K Tomblin
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Donald A Primerano
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Goran Boskovic
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
| | - Jun Fan
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Inderjit Mehmi
- Medical Oncology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
| | - Jackie Fletcher
- Department of Biology, West Virginia State University, Institute, WV 25112, USA.
| | - Nalini Santanam
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Estil Hurn
- Departments of Pharmacology, Physiology and Toxicology Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| | - Gary Z Morris
- Department of Science and Mathematics, Glenville State College, Glenville, WV 26351, USA.
| | - James Denvir
- Biochemistry and Microbiology and Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA.
| |
Collapse
|
41
|
Chokas AL, Bickford JS, Barilovits SJ, Rogers RJ, Qiu X, Newsom KJ, Beachy DE, Nick HS. A TEAD1/p65 complex regulates the eutherian-conserved MnSOD intronic enhancer, eRNA transcription and the innate immune response. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:1205-16. [PMID: 24953189 DOI: 10.1016/j.bbagrm.2014.06.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/11/2014] [Accepted: 06/13/2014] [Indexed: 12/21/2022]
Abstract
Manganese superoxide dismutase (MnSOD), a critical anti-oxidant enzyme, detoxifies the mitochondrial-derived reactive oxygen species, superoxide, elicited through normal respiration or the inflammatory response. Proinflammatory stimuli induce MnSOD gene expression through a eutherian-conserved, intronic enhancer element. We identified two prototypic enhancer binding proteins, TEAD1 and p65, that when co-expressed induce MnSOD expression comparable to pro-inflammatory stimuli. TEAD1 causes the nuclear sequestration of p65 leading to a novel TEAD1/p65 complex that associates with the intronic enhancer and is necessary for cytokine induction of MnSOD. Unlike typical NF-κB-responsive genes, the induction of MnSOD does not involve p50. Beyond MnSOD, the TEAD1/p65 complex regulates a subset of genes controlling the innate immune response that were previously viewed as solely NF-κB-dependent. We also identified an enhancer-derived RNA (eRNA) that is induced by either proinflammatory stimuli or the TEAD1/p65 complex, potentially linking the intronic enhancer to intra- and interchromosomal gene regulation through the inducible eRNA.
Collapse
Affiliation(s)
- Ann L Chokas
- Departments of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Justin S Bickford
- Departments of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sarah J Barilovits
- Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Richard J Rogers
- Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Anesthesiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Xiaolei Qiu
- Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kimberly J Newsom
- Departments of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dawn E Beachy
- Departments of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Harry S Nick
- Departments of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
42
|
Ongali B, Nicolakakis N, Tong XK, Aboulkassim T, Papadopoulos P, Rosa-Neto P, Lecrux C, Imboden H, Hamel E. Angiotensin II type 1 receptor blocker losartan prevents and rescues cerebrovascular, neuropathological and cognitive deficits in an Alzheimer's disease model. Neurobiol Dis 2014; 68:126-36. [PMID: 24807206 DOI: 10.1016/j.nbd.2014.04.018] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/17/2014] [Accepted: 04/27/2014] [Indexed: 11/18/2022] Open
Abstract
Angiotensin II (AngII) receptor blockers that bind selectively AngII type 1 (AT1) receptors may protect from Alzheimer's disease (AD). We studied the ability of the AT1 receptor antagonist losartan to cure or prevent AD hallmarks in aged (~18months at endpoint, 3months treatment) or adult (~12months at endpoint, 10months treatment) human amyloid precursor protein (APP) transgenic mice. We tested learning and memory with the Morris water maze, and evaluated neurometabolic and neurovascular coupling using [(18)F]fluoro-2-deoxy-D-glucose-PET and laser Doppler flowmetry responses to whisker stimulation. Cerebrovascular reactivity was assessed with on-line videomicroscopy. We measured protein levels of oxidative stress enzymes (superoxide dismutases SOD1, SOD2 and NADPH oxidase subunit p67phox), and quantified soluble and deposited amyloid-β (Aβ) peptide, glial fibrillary acidic protein (GFAP), AngII receptors AT1 and AT2, angiotensin IV receptor AT4, and cortical cholinergic innervation. In aged APP mice, losartan did not improve learning but it consolidated memory acquisition and recall, and rescued neurovascular and neurometabolic coupling and cerebrovascular dilatory capacity. Losartan normalized cerebrovascular p67phox and SOD2 protein levels and up-regulated those of SOD1. Losartan attenuated astrogliosis, normalized AT1 and AT4 receptor levels, but failed to rescue the cholinergic deficit and the Aβ pathology. Given preventively, losartan protected cognitive function, cerebrovascular reactivity, and AT4 receptor levels. Like in aged APP mice, these benefits occurred without a decrease in soluble Aβ species or plaque load. We conclude that losartan exerts potent preventive and restorative effects on AD hallmarks, possibly by mitigating AT1-initiated oxidative stress and normalizing memory-related AT4 receptors.
Collapse
Affiliation(s)
- Brice Ongali
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Nektaria Nicolakakis
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Tahar Aboulkassim
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | | | - Pedro Rosa-Neto
- Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Douglas Hospital Research Centre, McGill University, Montréal, QC H3A 2B4, Canada
| | - Clotilde Lecrux
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Hans Imboden
- Institute of Cell Biology, University of Bern, Switzerland
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada.
| |
Collapse
|
43
|
Abstract
SIGNIFICANCE Inflammation and immunity can be associated with varying degrees of heme release from hemoproteins, eventually leading to cellular and tissue iron (Fe) overload, oxidative stress, and tissue damage. Presumably, these deleterious effects contribute to the pathogenesis of systemic infections. RECENT ADVANCES Heme release from hemoglobin sensitizes parenchyma cells to undergo programmed cell death in response to proinflammatory cytokines, such as tumor necrosis factor. This cytotoxic effect is driven by a mechanism involving intracellular accumulation of free radicals, which sustain the activation of the c-Jun N-terminal kinase (JNK) signaling transduction pathway. While heme catabolism by heme oxygenase-1 (HO-1) prevents programmed cell death, this cytoprotective effect requires the co-expression of ferritin H (heart/heavy) chain (FTH), which controls the pro-oxidant effect of labile Fe released from the protoporphyrin IX ring of heme. This antioxidant effect of FTH restrains JNK activation, whereas JNK activation inhibits FTH expression, a cross talk that controls metabolic adaptation to cellular Fe overload associated with systemic infections. CRITICAL ISSUES AND FUTURE DIRECTIONS Identification and characterization of the mechanisms via which FTH provides metabolic adaptation to tissue Fe overload should provide valuable information to our current understanding of the pathogenesis of systemic infections as well as other immune-mediated inflammatory diseases.
Collapse
|
44
|
Kucinska M, Piotrowska H, Luczak MW, Mikula-Pietrasik J, Ksiazek K, Wozniak M, Wierzchowski M, Dudka J, Jäger W, Murias M. Effects of hydroxylated resveratrol analogs on oxidative stress and cancer cells death in human acute T cell leukemia cell line. Chem Biol Interact 2014; 209:96-110. [DOI: 10.1016/j.cbi.2013.12.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/13/2013] [Accepted: 12/20/2013] [Indexed: 01/13/2023]
|
45
|
Krautbauer S, Eisinger K, Neumeier M, Hader Y, Buettner R, Schmid PM, Aslanidis C, Buechler C. Free fatty acids, lipopolysaccharide and IL-1α induce adipocyte manganese superoxide dismutase which is increased in visceral adipose tissues of obese rodents. PLoS One 2014; 9:e86866. [PMID: 24475187 PMCID: PMC3901719 DOI: 10.1371/journal.pone.0086866] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 12/17/2013] [Indexed: 12/21/2022] Open
Abstract
Excess fat storage in adipocytes is associated with increased generation of reactive oxygen species (ROS) and impaired activity of antioxidant mechanisms. Manganese superoxide dismutase (MnSOD) is a mitochondrial enzyme involved in detoxification of ROS, and objective of the current study is to analyze expression and regulation of MnSOD in obesity. MnSOD is increased in visceral but not subcutaneous fat depots of rodents kept on high fat diets (HFD) and ob/ob mice. MnSOD is elevated in visceral adipocytes of fat fed mice and exposure of differentiating 3T3-L1 cells to lipopolysaccharide, IL-1α, saturated, monounsaturated and polyunsaturated free fatty acids (FFA) upregulates its level. FFA do not alter cytochrome oxidase 4 arguing against overall induction of mitochondrial enzymes. Upregulation of MnSOD in fat loaded cells is not mediated by IL-6, TNF or sterol regulatory element binding protein 2 which are induced in these cells. MnSOD is similarly abundant in perirenal fat of Zucker diabetic rats and non-diabetic animals with similar body weight and glucose has no effect on MnSOD in 3T3-L1 cells. To evaluate whether MnSOD affects adipocyte fat storage, MnSOD was knocked-down in adipocytes for the last three days of differentiation and in mature adipocytes. Knock-down of MnSOD does neither alter lipid storage nor viability of these cells. Heme oxygenase-1 which is induced upon oxidative stress is not altered while antioxidative capacity of the cells is modestly reduced. Current data show that inflammation and excess triglyceride storage raise adipocyte MnSOD which is induced in epididymal adipocytes in obesity.
Collapse
Affiliation(s)
- Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Markus Neumeier
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Yvonne Hader
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Roland Buettner
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
| | - Peter M. Schmid
- Department of Internal Medicine II, Regensburg University Hospital, Regensburg, Germany
| | - Charalampos Aslanidis
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany
- * E-mail:
| |
Collapse
|
46
|
D’Alessandro A, Zolla L. The SODyssey: superoxide dismutases from biochemistry, through proteomics, to oxidative stress, aging and nutraceuticals. Expert Rev Proteomics 2014; 8:405-21. [DOI: 10.1586/epr.11.13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
47
|
Laddha NC, Dwivedi M, Gani AR, Shajil EM, Begum R. Involvement of superoxide dismutase isoenzymes and their genetic variants in progression of and higher susceptibility to vitiligo. Free Radic Biol Med 2013; 65:1110-1125. [PMID: 24036105 DOI: 10.1016/j.freeradbiomed.2013.08.189] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 08/06/2013] [Accepted: 08/29/2013] [Indexed: 02/07/2023]
Abstract
Oxidative stress has been implicated as the initial triggering event in vitiligo pathogenesis leading to melanocyte destruction. Here, we report a significant increase in oxidative stress in vitiligo patients as evidenced by high lipid peroxidation levels suggesting an imbalance in the antioxidant enzyme system as reported in our previous studies. This study examined the role of the enzymatic antioxidant SOD, which converts the pro-oxidant superoxide into H2O2, in vitiligo pathogenesis. The activity of three isoforms of SOD, i.e., SOD1, SOD2, and SOD3, was significantly higher in vitiligo patients. To identify the underlying mechanism for the increase in activities of SOD isoforms, we explored the SOD1, SOD2, and SOD3 genes for their genetic variations and transcript levels. The SOD2 Thr58Ile (rs35289490) and Leu84Phe (rs11575993) polymorphisms were significantly associated with vitiligo patients, and the Val16Ala (rs4880) polymorphism was associated with active vitiligo patients. Interestingly, SOD2 activity was contributed by these polymorphisms along with its increase in transcript levels in patients. SOD3 activity was associated with the Arg213Gly (rs8192291) polymorphism. The SOD3 transcript levels were also increased in patients, which might contribute to the increased SOD3 activity. However, we could not establish the genotype-phenotype correlation for SOD1 as we could not detect any novel or reported SNPs in SOD1. In addition, both transcript and protein levels of SOD1 were unchanged between patients and controls, though SOD1 activity was increased in patients. Activities of SOD isoforms also correlated with progression of the disease as the activity was higher in active cases of vitiligo compared to stable cases. Here, we report that SOD2 and SOD3 polymorphisms may be genetic risk factors for susceptibility and progression of vitiligo and hence the genetic makeup of an individual may form a basis for the effective treatment of the disease. Overall, our results suggest that increased activity of SOD isoforms under the influence of genetic factors may lead to accumulation of H2O2 in cytoplasmic, mitochondrial, and extracellular compartments resulting in oxidative damage to the melanocytes.
Collapse
Affiliation(s)
- Naresh C Laddha
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002, India
| | - Mitesh Dwivedi
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002, India
| | - Amina R Gani
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002, India
| | - E M Shajil
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002, India.
| |
Collapse
|
48
|
Laddha NC, Dwivedi M, Gani AR, Mansuri MS, Begum R. Tumor necrosis factor B (TNFB) genetic variants and its increased expression are associated with vitiligo susceptibility. PLoS One 2013; 8:e81736. [PMID: 24312346 PMCID: PMC3842287 DOI: 10.1371/journal.pone.0081736] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 10/23/2013] [Indexed: 02/07/2023] Open
Abstract
Genetic polymorphisms in TNFB are involved in the regulation of its expression and are found to be associated with various autoimmune diseases. The aim of the present study was to determine whether TNFB +252A/G (rs909253) and exon 3 C/A (rs1041981) polymorphisms are associated with vitiligo susceptibility, and expression of TNFB and ICAM1 affects the disease onset and progression. We have earlier reported the role of TNFA in autoimmune pathogenesis of vitiligo, and we now show the involvement of TNFB in vitiligo pathogenesis. The two polymorphisms investigated in the TNFB were in strong linkage disequilibrium and significantly associated with vitiligo. TNFB and ICAM1 transcripts were significantly increased in patients compared to controls. Active vitiligo patients showed significant increase in TNFB transcripts compared to stable vitiligo. The genotype-phenotype analysis revealed that TNFB expression levels were higher in patients with GG and AA genotypes as compared to controls. Patients with the early age of onset and female patients showed higher TNFB and ICAM1 expression. Overall, our findings suggest that the increased TNFB transcript levels in vitiligo patients could result, at least in part, from variations at the genetic level which in turn leads to increased ICAM1 expression. For the first time, we show that TNFB +252A/G and exon 3 C/A polymorphisms are associated with vitiligo susceptibility and influence the TNFB and ICAM1 expression. Moreover, the study also emphasizes influence of TNFB and ICAM1 on the disease progression, onset and gender bias for developing vitiligo.
Collapse
Affiliation(s)
- Naresh C. Laddha
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Mitesh Dwivedi
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Amina R. Gani
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Mohmmad Shoab Mansuri
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
49
|
Pozniak PD, White MK, Khalili K. TNF-α/NF-κB signaling in the CNS: possible connection to EPHB2. J Neuroimmune Pharmacol 2013; 9:133-41. [PMID: 24277482 DOI: 10.1007/s11481-013-9517-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/13/2013] [Indexed: 12/27/2022]
Abstract
Tumor necrosis factor-alpha, TNF-α, is a cytokine that is a well-known factor in multiple disease conditions and is recognized for its major role in central nervous system signaling. TNF-α signaling is most commonly associated with neurotoxicity, but in some conditions it has been found to be neuroprotective. TNF-α has long been known to induce nuclear factor-kappa B, NF-κB, signaling by, in most cases, translocating the p65 (RelA) DNA binding factor to the nucleus. p65 is a key member of NF-κB, which is well established as a family of transcription factors that regulates many signaling events, including growth and process development, in neuronal cell populations. NF-κB has been shown to affect both the receiving aspect of neuronal signaling events in dendritic development as well as the sending of neuronal signals in axonal development. In both cases, NK-κB functions as a promoter and/or inhibitor of growth, depending on the environmental conditions and signaling cascade. In addition, NF-κB is involved in memory formation or neurogenesis, depending on the region of the brain in which the signaling occurs. The ephrin (Eph) receptor family represents a subfamily of receptor tyrosine kinases, RTKs, which received much attention due to its potential involvement in neuronal cell health and function. There are two subsets of ephrin receptors, Eph A and Eph B, each with distinct functions in cardiovascular and skeletal development and axon guidance and synaptic plasticity. The presence of multiple binding sites for NF-κB within the regulatory region of EphB2 gene and its potential regulation by NF-κB pathway suggests that TNF-α may modulate EphB2 via NF-κB and that this may contribute to the neuroprotective activity of TNF-α.
Collapse
Affiliation(s)
- Paul D Pozniak
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Room 741 MERB, 3500N. Broad Street, Philadelphia, PA, 19140, USA
| | | | | |
Collapse
|
50
|
Wu S, Xu X, Zhao S, Shen F, Chen J. Evaluation of phenanthrene toxicity on earthworm (Eisenia fetida): an ecotoxicoproteomics approach. CHEMOSPHERE 2013; 93:963-971. [PMID: 23856470 DOI: 10.1016/j.chemosphere.2013.05.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 05/23/2013] [Accepted: 05/25/2013] [Indexed: 06/02/2023]
Abstract
The goal of this study was to identify promising new biomarkers of phenanthrene by identifying differentially expressed proteins in Eisenia fetida after exposure to phenanthrene. Extracts of earthworm epithelium collected at days 2, 7, 14, and 28 after phenanthrene exposure were analyzed by two dimensional electrophoresis (2-DE) and quantitative image analysis. Comparing the intensity of protein spots, 36 upregulated proteins and 45 downregulated proteins were found. Some of the downregulated and upregulated proteins were verified by MALDI-TOF/TOF-MS and database searching. Downregulated proteins in response to phenanthrene exposure were involved in glycolysis, energy metabolism, chaperones, proteolysis, protein folding and electron transport. In contrast, oxidation reduction, oxygen transport, defense systems response to pollutant, protein biosynthesis and fatty acid biosynthesis were upregulated in phenanthrene-treated E. fetida. In addition, ATP synthase b subunit, lysenin-related protein 2, lombricine kinase, glyceraldehyde 3-phosphate dehydrogenase, actinbinding protein, and extracellular globin-4 seem to be potential biomarkers since these biomarker were able to low levels (2.5 mg kg(-1)) of phenanthrene. Our study provides a functional profile of the phenanthrene-responsive proteins in earthworms. The variable levels and trends in these spots could play a potential role as novel biomarkers for monitoring the levels of phenanthrene contamination in soil ecosystems.
Collapse
Affiliation(s)
- Shijin Wu
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, People's Republic of China
| | | | | | | | | |
Collapse
|