1
|
Tang Y, Guo T, Wang X, Li C, Zhang X, Zhang J. Cyclodextrin-Derived Macromolecular Therapies for Inflammatory Diseases. Macromol Biosci 2025:e2400637. [PMID: 40271896 DOI: 10.1002/mabi.202400637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Inflammation is an essential physiological defense mechanism against harmful stimuli, yet dysregulated inflammatory responses are closely associated with the pathogenesis of numerous acute and chronic diseases. Recent advances highlight the remarkable anti-inflammatory potential of bioactive macromolecules, particularly cyclodextrins (CDs) and their engineered derivatives, which are emerging as promising therapeutic agents. This review systematically introduces different CDs and CD-derived macromolecules that demonstrate anti-inflammatory properties, with emphasis on their molecular mechanisms of action. Native CDs exhibit direct therapeutic effects through host-guest interactions, enabling selective sequestration of pathogenic components such as cholesterol crystals and proteins that drive inflammatory cascades. Moreover, chemically modified CD derivatives incorporating functional groups demonstrate enhanced capabilities in neutralizing inflammatory mediators and modulating immune cell responses. This work further discusses the expanding therapeutic applications of these macromolecules across diverse inflammatory conditions, ranging from acute tissue injuries to chronic autoimmune disorders. Finally, this work critically analyzes the crucial challenges and emerging opportunities in translating CD-based macromolecular therapies into clinical practice, addressing key considerations in biocompatibility, targeted delivery, and therapeutic efficacy optimization.
Collapse
Affiliation(s)
- Yige Tang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xuanran Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, China
| |
Collapse
|
2
|
Hipper E, Diederichs T, Kaiser W, Lehmann F, Buske J, Hinderberger D, Garidel P. Visible light triggers the formation of reactive oxygen species in monoclonal antibody formulations. Int J Pharm 2024; 661:124392. [PMID: 38942184 DOI: 10.1016/j.ijpharm.2024.124392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Most monoclonal antibody formulations require the presence of a surfactant, such as polysorbate, to ensure protein stability. The presence of high concentrations of polysorbate have been shown to enhance photooxidation of certain protein drug products when exposed to visible light. The current literature, however, suggest that photooxidation of polysorbate only occurs when exposed to visible light in combination with UVA light. This is probable as peroxides present in polysorbate solutions can be cleaved homolytically in the UVA region. In the visible region, photooxidation is not expected to occur as cleavage of peroxides is not expected at these wavelengths. This report presents findings suggesting that the presence of one or more photosensitiser(s) in polysorbate must be a cause and is required to catalyse the aerobic oxidation of polysorbate solutions upon exposure to visible light. Our investigation aimed to clarify the mechanism(s) of polysorbate photooxidation and explore the kinetics and the identity of the generated radicals and their impact on monoclonal antibody (mAb) degradation. Our study reveals that when polysorbate solutions are exposed to visible light between 400 - 800 nm in the absence of proteins, discolouration, radical formation, and oxygen depletion occur. We discuss the initial formation of reactive species, most likely occurring directly after reaction of molecular oxygen, with the presence of a triplet state photosensitiser, which is generated by intersystem crossing of the excited singlet state. When comparing the photooxidation of PS20 and PS80 in varying quality grades, we propose that singlet oxygen possesses potential for reacting with unsaturated fatty acids in PS80HP, however, PS20HP itself exhibited no measurable oxidation under the tested conditions. The study's final part delves into the photooxidation behaviour of different PS grades, examining its influence on the integrity of a mAb in the formulation. Finally, we examined the effect of photooxidation on the integrity of monoclonal antibodies. Our findings show that the exposure to visible light in polysorbate-containing mAb solutions at high PS concentrations of 4 mg·ml-1 results in increased monoclonal antibody degradation, highlighting the need for cautious evaluation of the correct PS concentration to stabilise protein therapeutics.
Collapse
Affiliation(s)
- Elena Hipper
- Institute of Chemistry, Martin-Luther-University Halle-Wittenberg, Von-Danckelmann-Platz 4, 06120 Halle, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Tim Diederichs
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Wolfgang Kaiser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Florian Lehmann
- Institute of Chemistry, Martin-Luther-University Halle-Wittenberg, Von-Danckelmann-Platz 4, 06120 Halle, Germany
| | - Julia Buske
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany
| | - Dariush Hinderberger
- Institute of Chemistry, Martin-Luther-University Halle-Wittenberg, Von-Danckelmann-Platz 4, 06120 Halle, Germany
| | - Patrick Garidel
- Institute of Chemistry, Martin-Luther-University Halle-Wittenberg, Von-Danckelmann-Platz 4, 06120 Halle, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, Birkendorfer Straße 65, 88397 Biberach an der Riss, Germany.
| |
Collapse
|
3
|
Chidambaram SB, Anand N, Varma SR, Ramamurthy S, Vichitra C, Sharma A, Mahalakshmi AM, Essa MM. Superoxide dismutase and neurological disorders. IBRO Neurosci Rep 2024; 16:373-394. [PMID: 39007083 PMCID: PMC11240301 DOI: 10.1016/j.ibneur.2023.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/21/2023] [Indexed: 07/16/2024] Open
Abstract
Superoxide dismutase (SOD) is a common antioxidant enzyme found majorly in living cells. The main physiological role of SOD is detoxification and maintain the redox balance, acts as a first line of defence against Reactive nitrogen species (RNS), Reactive oxygen species (ROS), and other such potentially hazardous molecules. SOD catalyses the conversion of superoxide anion free radicals (O 2 -.) into molecular oxygen (O 2) and hydrogen peroxide (H 2O 2) in the cells. Superoxide dismutases (SODs) are expressed in neurons and glial cells throughout the CNS both intracellularly and extracellularly. Endogenous oxidative stress (OS) linked with enlarged production of reactive oxygen metabolites (ROMs), inflammation, deregulation of redox balance, mitochondrial dysfunction and bioenergetic crisis are found to be prerequisite for neuronal loss in neurological diseases. Clinical and genetic studies indicate a direct correlation between mutations in SOD gene and neurodegenerative diseases, like Amyotrophic Lateral Sclerosis (ALS), Huntington's disease (HD), Parkinson's Disease (PD) and Alzheimer's Disease (AD). Therefore, inhibitors of OS are considered as an optimistic approach to prevent neuronal loss. SOD mimetics like Metalloporphyrin Mn (II)-cyclic polyamines, Nitroxides and Mn (III)- Salen complexes are designed and used as therapeutic extensively in the treatment of neurological disorders. SODs and SOD mimetics are promising future therapeutics in the field of various diseases with OS-mediated pathology.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department of Pharmacology, American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Antigua, Antigua and Barbuda
| | - Sudhir Rama Varma
- Department of Clinical Sciences, College of Dentistry, Ajman University, 346 Ajman, the United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University, 346 Ajman, the United Arab Emirates
| | - Srinivasan Ramamurthy
- College of Pharmacy & Health Sciences, University of Science and Technology of Fujairah, 2202 Fujairah, the United Arab Emirates
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Ambika Sharma
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
4
|
Qi C, Bujaroski RS, Baell J, Zheng X. Kinases in cerebral cavernous malformations: Pathogenesis and therapeutic targets. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119488. [PMID: 37209718 DOI: 10.1016/j.bbamcr.2023.119488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
Cerebral cavernous malformations (CCMs) are low-flow, hemorrhagic vascular lesions of the central nervous system of genetic origin, which can cause stroke-like symptoms and seizures. From the identification of CCM1, CCM2 and CCM3 as genes related to disease progression, molecular and cellular mechanisms for CCM pathogenesis have been established and the search for potential drugs to target CCM has begun. Broadly speaking, kinases are the major group signaling in CCM pathogenesis. These include the MEKK3/MEK5/ERK5 cascade, Rho/Rock signaling, CCM3/GCKIII signaling, PI3K/mTOR signaling, and others. Since the discovery of Rho/Rock in CCM pathogenesis, inhibitors for Rho signaling and subsequently other components in CCM signaling were discovered and applied in preclinical and clinical trials to ameliorate CCM progression. This review discusses the general aspects of CCM disease, kinase-mediated signaling in CCM pathogenesis and the current state of potential treatment options for CCM. It is suggested that kinase target drug development in the context of CCM might facilitate and meet the unmet requirement - a non-surgical option for CCM disease.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, China
| | - Richard Sean Bujaroski
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Australian Translational Medicinal Chemistry Facility (ATMCF), Monash University, Parkville, Victoria, Australia
| | - Jonathan Baell
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, China
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, China.
| |
Collapse
|
5
|
Luan Y, Luo Y, Deng M. New advances in Nrf2-mediated analgesic drugs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154598. [PMID: 36603339 DOI: 10.1016/j.phymed.2022.154598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/24/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Oxidative stress is an inevitable process that occurs during life activities, and it can participate in the development of inflammation. Although great progress has been made according to research examining analgesic drugs and therapies, there remains a need to develop new analgesic drugs to fill certain gaps in both the experimental and clinical space. PURPOSE This review reports the research and preclinical progress of this class of analgesics by summarizing known nuclear factor E-2-related factor-2 (Nrf2) pathway-modulating substances. STUDY DESIGN We searched and reported experiments that intervene in the Nrf2 pathway and its various upstream and downstream molecules for analgesic therapy. METHODS The medical literature database (PubMed) was searched for experimental studies examining the reduction of pain in animals through the Nrf2 pathway, the research methods were analyzed, and the pathways were classified and reported according to the pathway of these experimental interventions. RESULTS Humans have identified a variety of substances that can fight pain by regulating the expression of Nrf2 and its upstream and downstream pathways. CONCLUSION The Nrf2 pathway exerts anti-inflammatory activity by regulating oxidative stress, thereby playing a role in the fight against pain.
Collapse
Affiliation(s)
- Yifan Luan
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410013, China
| | - Yaping Luo
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410013, China
| | - Meichun Deng
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
6
|
Pothion H, Lihrmann I, Duclos C, Riou G, Cartier D, Boukhzar L, Lefranc B, Leprince J, Guérout N, Marie JP, Anouar Y. The SELENOT mimetic PSELT promotes nerve regeneration by increasing axonal myelination in a facial nerve injury model in female rats. J Neurosci Res 2022; 100:1721-1731. [PMID: 35730417 PMCID: PMC9545325 DOI: 10.1002/jnr.25098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 03/17/2022] [Accepted: 06/06/2022] [Indexed: 01/31/2023]
Abstract
Peripheral nerve injury (PNI) is frequent and many patients suffer lifelong disabilities in severe cases. Although the peripheral nervous system is able to regenerate, its potential is limited. In this study, we tested in a nerve regeneration model in rat the potential beneficial effect of a short mimetic peptide, named PSELT, which derives from SELENOT, an essential thioredoxin-like selenoprotein endowed with neuroprotective and antioxidant activities. For this purpose, the right facial nerve of female Long-Evans rats was axotomized then bridged with a free femoral vein interposition graft. PSELT (1 μM) was injected into the vein immediately and 48 h after the injury, and the effects observed were compared to those found after an end-to-end suture used as a gold standard treatment. Whisking behavior, electrophysiological potential, and histological analyses were performed 3 months after injury to determine the effects of these treatments. These analyses revealed that PSELT-treated animals exhibit a better motor recovery in terms of protraction amplitude and velocity of vibrissae compared to control and end-sutured nerve animal groups. Moreover, administration of PSELT following injury enhanced muscle innervation, axonal elongation, and myelination of newly formed nerve fibers. Altogether, these results indicate that a PSELT-based treatment is sufficient to enhance facial nerve myelination and regeneration and could represent a new therapeutic tool to treat PNI.
Collapse
Affiliation(s)
- Hugo Pothion
- Normandie Univ, UNIROUEN, INSERM U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France.,Normandie Univ, UNIROUEN, UR 3830, Groupe de Recherche sur l'Handicap Ventilatoire et Neurologique, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France.,Fédération Hospitalo-Universitaire (FHU) Surface, Rouen, France
| | - Isabelle Lihrmann
- Normandie Univ, UNIROUEN, INSERM U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Celia Duclos
- Normandie Univ, UNIROUEN, UR 3830, Groupe de Recherche sur l'Handicap Ventilatoire et Neurologique, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Gaëtan Riou
- Normandie Univ, UNIROUEN, INSERM U1234, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Dorthe Cartier
- Normandie Univ, UNIROUEN, INSERM U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Loubna Boukhzar
- Normandie Univ, UNIROUEN, INSERM U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Benjamin Lefranc
- Normandie Univ, UNIROUEN, INSERM U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France.,Normandie Univ, UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Jérôme Leprince
- Normandie Univ, UNIROUEN, INSERM U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France.,Normandie Univ, UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Nicolas Guérout
- Normandie Univ, UNIROUEN, UR 3830, Groupe de Recherche sur l'Handicap Ventilatoire et Neurologique, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France.,Fédération Hospitalo-Universitaire (FHU) Surface, Rouen, France
| | - Jean-Paul Marie
- Normandie Univ, UNIROUEN, UR 3830, Groupe de Recherche sur l'Handicap Ventilatoire et Neurologique, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France.,Fédération Hospitalo-Universitaire (FHU) Surface, Rouen, France.,Otorhinolaryngology and Head Neck Surgery Department, Rouen University Hospital, Rouen, France
| | - Youssef Anouar
- Normandie Univ, UNIROUEN, INSERM U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France.,Fédération Hospitalo-Universitaire (FHU) Surface, Rouen, France
| |
Collapse
|
7
|
Chacko KM, Nouri MZ, Schramm WC, Malik Z, Liu LP, Denslow ND, Alli AA. Tempol Alters Urinary Extracellular Vesicle Lipid Content and Release While Reducing Blood Pressure during the Development of Salt-Sensitive Hypertension. Biomolecules 2021; 11:biom11121804. [PMID: 34944449 PMCID: PMC8699083 DOI: 10.3390/biom11121804] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/27/2021] [Indexed: 12/11/2022] Open
Abstract
Salt-sensitive hypertension resulting from an increase in blood pressure after high dietary salt intake is associated with an increase in the production of reactive oxygen species (ROS). ROS are known to increase the activity of the epithelial sodium channel (ENaC), and therefore, they have an indirect effect on sodium retention and increasing blood pressure. Extracellular vesicles (EVs) carry various molecules including proteins, microRNAs, and lipids and play a role in intercellular communication and intracellular signaling in health and disease. We investigated changes in EV lipids, urinary electrolytes, osmolality, blood pressure, and expression of renal ENaC and its adaptor protein, MARCKS/MARCKS Like Protein 1 (MLP1) after administration of the antioxidant Tempol in salt-sensitive hypertensive 129Sv mice. Our results show Tempol infusion reduces systolic blood pressure and protein expression of the alpha subunit of ENaC and MARCKS in the kidney cortex of hypertensive 129Sv mice. Our lipidomic data show an enrichment of diacylglycerols and monoacylglycerols and reduction in ceramides, dihydroceramides, and triacylglycerols in urinary EVs from these mice after Tempol treatment. These data will provide insight into our understanding of mechanisms involving strategies aimed to inhibit ROS to alleviate salt-sensitive hypertension.
Collapse
Affiliation(s)
- Kevin M. Chacko
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA; (K.M.C.); (W.C.S.); (Z.M.); (L.P.L.)
| | - Mohammad-Zaman Nouri
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA; (M.-Z.N.); (N.D.D.)
| | - Whitney C. Schramm
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA; (K.M.C.); (W.C.S.); (Z.M.); (L.P.L.)
| | - Zeeshan Malik
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA; (K.M.C.); (W.C.S.); (Z.M.); (L.P.L.)
| | - Lauren P. Liu
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA; (K.M.C.); (W.C.S.); (Z.M.); (L.P.L.)
| | - Nancy D. Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA; (M.-Z.N.); (N.D.D.)
| | - Abdel A. Alli
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA; (K.M.C.); (W.C.S.); (Z.M.); (L.P.L.)
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
- Correspondence: ; Tel.: +1-(352)-273-7877
| |
Collapse
|
8
|
Baby SM, Discala JF, Gruber R, Getsy PM, Cheng F, Damron DS, Lewis SJ. Tempol Reverses the Negative Effects of Morphine on Arterial Blood-Gas Chemistry and Tissue Oxygen Saturation in Freely-Moving Rats. Front Pharmacol 2021; 12:749084. [PMID: 34630119 PMCID: PMC8493249 DOI: 10.3389/fphar.2021.749084] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022] Open
Abstract
We have reported that pretreatment with the clinically approved superoxide dismutase mimetic, Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl), blunts the cardiorespiratory depressant responses elicited by a subsequent injection of fentanyl, in halothane-anesthetized rats. The objective of the present study was to determine whether Tempol is able to reverse the effects of morphine on arterial blood-gas (ABG) chemistry in freely-moving Sprague Dawley rats. The intravenous injection of morphine (10 mg/kg) elicited substantial decreases in pH, pO2 and sO2 that were accompanied by substantial increases in pCO2 and Alveolar-arterial gradient, which results in diminished gas-exchange within the lungs. Intravenous injection of a 60 mg/kg dose of Tempol 15 min after the injection of morphine caused minor improvements in pO2 and pCO2 but not in other ABG parameters. In contrast, the 100 mg/kg dose of Tempol caused an immediate and sustained reversal of the negative effects of morphine on arterial blood pH, pCO2, pO2, sO2 and Alveolar-arterial gradient. In other rats, we used pulse oximetry to determine that the 100 mg/kg dose of Tempol, but not the 60 mg/kg dose elicited a rapid and sustained reversal of the negative effects of morphine (10 mg/kg, IV) on tissue O2 saturation (SpO2). The injection of morphine caused a relatively minor fall in mean arterial blood pressure that was somewhat exacerbated by Tempol. These findings demonstrate that Tempol can reverse the negative effects of morphine on ABG chemistry in freely-moving rats paving the way of structure-activity and mechanisms of action studies with the host of Tempol analogues that are commercially available.
Collapse
Affiliation(s)
| | | | - Ryan Gruber
- Galleon Pharmaceuticals Inc, Horsham, PA, United states
| | - Paulina M Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United states
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United states
| | - Derek S Damron
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH, United states
| | - Stephen J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United states.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United states
| |
Collapse
|
9
|
Zhang Y, Tang J, Li N, Tao J, Zhang Y, Zhang Y, Ye Y, Zheng Q, Xu T, Liu Y, Zhang P, Li L, Li H, He Y, Su H, He Q, Sun M, Xu Z. Prenatal hypoxia induced ET BR activation and abnormal ROS signalling in pulmonary artery cells of rat offspring. Reprod Toxicol 2021; 105:91-100. [PMID: 34478853 DOI: 10.1016/j.reprotox.2021.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/24/2021] [Accepted: 08/29/2021] [Indexed: 12/22/2022]
Abstract
Pulmonary arterial hypertension is a progressive disorder characterized by remodeling and increased small pulmonary arteries resistance. Endothelin-1 (ET-1) was related to PAH and ET-1 receptors were up-regulated selectively in the lung when exposed to toxic factor hypoxia. However, the role of ET-1 signaling in the pathogenesis of prenatal hypoxia-induced pulmonary abnormalities remains to be elucidated. Pregnant rats were divided into prenatal hypoxia (10.5 % O2 from gestational day 4-21) and control group. Their three-month-old offspring male rats were tested for vascular functions and molecular analysis, DNA methylation was assessed for cellular hypoxia. Functional testing showed that ET-1-mediated vasoconstriction was enhanced, and the expressions of endothelin A receptor/B receptor (ETAR/ETBR), inositol 1,4,5-trisphosphate receptor, type 1, and the sensitivity of calcium channels were increased in the small pulmonary arteries following prenatal hypoxia. q-PCR and DHE staining showed that the expressions of NADPH oxidase 1/4 (Nox1/4) were up-regulated, along with the increased production of superoxide anion. Furthermore, superoxide anion promoted ET-1-mediated pulmonary artery contraction. In the pulmonary artery smooth muscle cell experiments, q-PCR, Western Blot, CCK8 and DHE staining showed that the expressions of ETBR, Nox1/4, and superoxide anion were increased by hypoxia, along with promoted cell proliferation. 2,2,6,6-Tetramethyl-1-piperidinyloxy reversed hypoxia-induced cell proliferation. ETBR antagonist BQ788 inhibited hypoxia-increased expressions of Nox1/4, superoxide anion production, and proliferation of cells. Moreover, methylation analysis indicated that hypoxia decreased the methylation levels of the ETBR promoter in the pulmonary artery smooth muscle cells. The results indicated that prenatal toxic factor hypoxia resulted in abnormal ETBR activation, which enhanced ET-1-mediated vasoconstriction of pulmonary arteries and pulmonary artery smooth muscle cell proliferation through ETBR/Nox1/4-derived ROS pathway.
Collapse
Affiliation(s)
- Yingying Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China; Wuxi Maternal & Child Health Hospital, Jiangsu, China
| | - Jiaqi Tang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Na Li
- Wuxi Maternal & Child Health Hospital, Jiangsu, China
| | - Jianying Tao
- Department of Obstetrics & Gynecology, Suzhou Municipal Hospital, Suzhou, China
| | - Yan Zhang
- Wuxi Maternal & Child Health Hospital, Jiangsu, China
| | - Yumeng Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yang Ye
- Wuxi Maternal & Child Health Hospital, Jiangsu, China
| | - Qiutong Zheng
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Ting Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yanping Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Pengjie Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Lingjun Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Huan Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yun He
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Hongyu Su
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Qinyuan He
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China; Wuxi Maternal & Child Health Hospital, Jiangsu, China.
| |
Collapse
|
10
|
Herb M, Gluschko A, Schramm M. Reactive Oxygen Species: Not Omnipresent but Important in Many Locations. Front Cell Dev Biol 2021; 9:716406. [PMID: 34557488 PMCID: PMC8452931 DOI: 10.3389/fcell.2021.716406] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023] Open
Abstract
Reactive oxygen species (ROS), such as the superoxide anion or hydrogen peroxide, have been established over decades of research as, on the one hand, important and versatile molecules involved in a plethora of homeostatic processes and, on the other hand, as inducers of damage, pathologies and diseases. Which effects ROS induce, strongly depends on the cell type and the source, amount, duration and location of ROS production. Similar to cellular pH and calcium levels, which are both strictly regulated and only altered by the cell when necessary, the redox balance of the cell is also tightly regulated, not only on the level of the whole cell but in every cellular compartment. However, a still widespread view present in the scientific community is that the location of ROS production is of no major importance and that ROS randomly diffuse from their cellular source of production throughout the whole cell and hit their redox-sensitive targets when passing by. Yet, evidence is growing that cells regulate ROS production and therefore their redox balance by strictly controlling ROS source activation as well as localization, amount and duration of ROS production. Hopefully, future studies in the field of redox biology will consider these factors and analyze cellular ROS more specifically in order to revise the view of ROS as freely flowing through the cell.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Alexander Gluschko
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Michael Schramm
- Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| |
Collapse
|
11
|
Baby S, Gruber R, Discala J, Puskovic V, Jose N, Cheng F, Jenkins M, Seckler J, Lewis S. Systemic Administration of Tempol Attenuates the Cardiorespiratory Depressant Effects of Fentanyl. Front Pharmacol 2021; 12:690407. [PMID: 34248639 PMCID: PMC8260831 DOI: 10.3389/fphar.2021.690407] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/11/2021] [Indexed: 11/30/2022] Open
Abstract
Fentanyl is a high-potency opioid receptor agonist that elicits profound analgesia and suppression of breathing in humans and animals. To date, there is limited evidence as to whether changes in oxidant stress are important factors in any of the actions of acutely administered fentanyl. This study determined whether the clinically approved superoxide dismutase mimetic, Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl), or a potent antioxidant, N-acetyl-L-cysteine methyl ester (L-NACme), modify the cardiorespiratory and analgesic actions of fentanyl. We examined whether the prior systemic injection of Tempol or L-NACme affects the cardiorespiratory and/or analgesic responses elicited by the subsequent injection of fentanyl in isoflurane-anesthetized and/or freely moving male Sprague-Dawley rats. Bolus injections of Tempol (25, 50 or 100 mg/kg, IV) elicited minor increases in frequency of breathing, tidal volume and minute ventilation. The ventilatory-depressant effects of fentanyl (5 μg/kg, IV) given 15 min later were dose-dependently inhibited by prior injections of Tempol. Tempol elicited dose-dependent and transient hypotension that had (except for the highest dose) resolved when fentanyl was injected. The hypotensive responses elicited by fentanyl were markedly blunted after Tempol pretreatment. The analgesic actions of fentanyl (25 μg/kg, IV) were not affected by Tempol (100 mg/kg, IV). L-NACme did not modify any of the effects of fentanyl. We conclude that prior administration of Tempol attenuates the cardiorespiratory actions of fentanyl without affecting the analgesic effects of this potent opioid. As such, Tempol may not directly affect opioid-receptors that elicit the effects of fentanyl. Whether, the effects of Tempol are solely due to alterations in oxidative stress is in doubt since the powerful antioxidant, L-NACme, did not affect fentanyl-induced suppression of breathing.
Collapse
Affiliation(s)
- Santhosh Baby
- Galleon Pharmaceuticals, Inc., Horsham, PA, United States
| | - Ryan Gruber
- Galleon Pharmaceuticals, Inc., Horsham, PA, United States
| | - Joseph Discala
- Galleon Pharmaceuticals, Inc., Horsham, PA, United States
| | | | - Nijo Jose
- Department of Radiotherapy and Oncology, Kasturba Medical College, Manipal, India
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Michael Jenkins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States.,Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - James Seckler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Stephen Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
12
|
Martin NJ, Chami B, Vallejo A, Mojadadi AA, Witting PK, Ahmad G. Efficacy of the Piperidine Nitroxide 4-MethoxyTEMPO in Ameliorating Serum Amyloid A-Mediated Vascular Inflammation. Int J Mol Sci 2021; 22:ijms22094549. [PMID: 33925294 PMCID: PMC8123591 DOI: 10.3390/ijms22094549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 01/24/2023] Open
Abstract
Intracellular redox imbalance in endothelial cells (EC) can lead to endothelial dysfunction, which underpins cardiovascular diseases (CVD). The acute phase serum amyloid A (SAA) elicits inflammation through stimulating production of reactive oxygen species (ROS). The cyclic nitroxide 4-MethoxyTEMPO (4-MetT) is a superoxide dismutase mimetic that suppresses oxidant formation and inflammation. The aim of this study was to investigate whether 4-MetT inhibits SAA-mediated activation of cultured primary human aortic EC (HAEC). Co-incubating cells with 4-MetT inhibited SAA-mediated increases in adhesion molecules (VCAM-1, ICAM-1, E-selectin, and JAM-C). Pre-treatment of cells with 4-MetT mitigated SAA-mediated increases in transcriptionally activated NF-κB-p65 and P120 Catenin (a stabilizer of Cadherin expression). Mitochondrial respiration and ROS generation (mtROS) were adversely affected by SAA with decreased respiratory reserve capacity, elevated maximal respiration and proton leakage all characteristic of SAA-treated HAEC. This altered respiration manifested as a loss of mitochondrial membrane potential (confirmed by a decrease in TMRM fluorescence), and increased mtROS production as assessed with MitoSox Red. These SAA-linked impacts on mitochondria were mitigated by 4-MetT resulting in restoration of HAEC nitric oxide bioavailability as confirmed by assessing cyclic guanosine monophosphate (cGMP) levels. Thus, 4-MetT ameliorates SAA-mediated endothelial dysfunction through normalising EC redox homeostasis. Subject to further validation in in vivo settings; these outcomes suggest its potential as a therapeutic in the setting of cardiovascular pathologies where elevated SAA and endothelial dysfunction is linked to enhanced CVD.
Collapse
|
13
|
Rosa AC, Corsi D, Cavi N, Bruni N, Dosio F. Superoxide Dismutase Administration: A Review of Proposed Human Uses. Molecules 2021; 26:1844. [PMID: 33805942 PMCID: PMC8037464 DOI: 10.3390/molecules26071844] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Superoxide dismutases (SODs) are metalloenzymes that play a major role in antioxidant defense against oxidative stress in the body. SOD supplementation may therefore trigger the endogenous antioxidant machinery for the neutralization of free-radical excess and be used in a variety of pathological settings. This paper aimed to provide an extensive review of the possible uses of SODs in a range of pathological settings, as well as describe the current pitfalls and the delivery strategies that are in development to solve bioavailability issues. We carried out a PubMed query, using the keywords "SOD", "SOD mimetics", "SOD supplementation", which included papers published in the English language, between 2012 and 2020, on the potential therapeutic applications of SODs, including detoxification strategies. As highlighted in this paper, it can be argued that the generic antioxidant effects of SODs are beneficial under all tested conditions, from ocular and cardiovascular diseases to neurodegenerative disorders and metabolic diseases, including diabetes and its complications and obesity. However, it must be underlined that clinical evidence for its efficacy is limited and consequently, this efficacy is currently far from being demonstrated.
Collapse
Affiliation(s)
- Arianna Carolina Rosa
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Daniele Corsi
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Niccolò Cavi
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Natascia Bruni
- Istituto Farmaceutico Candioli, Strada Comunale di None, 1, 10092 Beinasco, Italy;
| | - Franco Dosio
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| |
Collapse
|
14
|
Functions of ROS in Macrophages and Antimicrobial Immunity. Antioxidants (Basel) 2021; 10:antiox10020313. [PMID: 33669824 PMCID: PMC7923022 DOI: 10.3390/antiox10020313] [Citation(s) in RCA: 322] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are a chemically defined group of reactive molecules derived from molecular oxygen. ROS are involved in a plethora of processes in cells in all domains of life, ranging from bacteria, plants and animals, including humans. The importance of ROS for macrophage-mediated immunity is unquestioned. Their functions comprise direct antimicrobial activity against bacteria and parasites as well as redox-regulation of immune signaling and induction of inflammasome activation. However, only a few studies have performed in-depth ROS analyses and even fewer have identified the precise redox-regulated target molecules. In this review, we will give a brief introduction to ROS and their sources in macrophages, summarize the versatile roles of ROS in direct and indirect antimicrobial immune defense, and provide an overview of commonly used ROS probes, scavengers and inhibitors.
Collapse
|
15
|
Nagayasu M, Imanaka S, Kimura M, Maruyama S, Kobayashi H. Nonhormonal Treatment for Endometriosis Focusing on Redox Imbalance. Gynecol Obstet Invest 2021; 86:1-12. [PMID: 33395684 DOI: 10.1159/000512628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/26/2020] [Indexed: 11/19/2022]
Abstract
The aim of this review is to investigate the oxidant/antioxidant status and its regulatory mechanisms in patients with endometriosis and to summarize the antioxidant therapy as an alternative to hormonal therapy for endometriosis. Each keyword alone or in combination was used to search from PubMed and Embase by applying the filters of the title and the publication years between January 2000 and March 2020. Endometriosis is a chronic inflammatory disease characterized by repeated episodes of hemorrhage. Methemoglobin in repeated hemorrhage produces large amounts of superoxide anion via the autoxidation of hemoglobin. Excessive free-radical production causes redox imbalance, leading to inadequate antioxidant defenses and damage to endometrial cells, but may contribute to endometrial cell growth and survival through activation of various signaling pathways. In addition, to overcome excessive oxidative stress, estradiol participates in the induction of antioxidants such as superoxide dismutase in mitochondria. Several antioxidants that suppress free radicals may be effective in endometriosis-related pain. We searched for 23 compounds and natural substances that could reduce the pain caused by superoxide/reactive oxygen species in basic research and animal models. Next, we built a list of 16 drugs that were suggested to be effective against endometriosis other than hormone therapy in preclinical studies and clinical trials. Of the 23 and 16 drugs, 4 overlapping drugs could be potential candidates for clinically reducing endometriosis-related pain caused by superoxide anion/reactive oxygen species. These drugs include polyphenols (resveratrol and polydatin), dopamine agonists (cabergoline), and statins (simvastatin). However, no randomized controlled trials have evaluated the efficacy of these drugs. In conclusion, this review summarizes the following 2 points: superoxide anion generation by methemoglobin is enhanced in endometriosis, resulting in redox imbalance; and some compounds and natural substances that can suppress free radicals may be effective in endometriosis-related pain. Further randomized clinical trials based on larger series are mandatory to confirm the promising role of antioxidants in the nonhormonal management of endometriosis.
Collapse
Affiliation(s)
- Mika Nagayasu
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Shogo Imanaka
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan.,Ms.Clinic MayOne, Kashihara, Japan
| | - Mai Kimura
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Sachiyo Maruyama
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan, .,Ms.Clinic MayOne, Kashihara, Japan,
| |
Collapse
|
16
|
Hermes TDA, Mizobuti DS, da Rocha GL, da Silva HNM, Covatti C, Pereira ECL, Ferretti R, Minatel E. Tempol improves redox status in mdx dystrophic diaphragm muscle. Int J Exp Pathol 2020; 101:289-297. [PMID: 33098599 DOI: 10.1111/iep.12376] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/10/2020] [Accepted: 08/23/2020] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress is a critical element in relationship to the pathophysiology of Duchenne muscular dystrophy (DMD). In the mice the diaphragm (DIA) is most resembles the dystrophic human pathology. In this study we have evaluated the consequences of a synthetic antioxidant (tempol) on oxidative stress parameters in the DIA muscle of mdx mice. The mdx mice were separated into two groups: mdx, the control group receiving intraperitoneal (i.p.) injections of saline solution (100 µL), and mdxT, the treated group receiving i.p. injections of tempol (100 mg/kg). The tempol-treated group showed reduced oxidative stress markers, decreasing the dihydroethidium reaction (DHE) area; autofluorescent lipofuscin granules; and 4-hydroxynonenal (4-HNE)-protein adduct levels. DIA muscle of mdx mice. At the same time, the manganese-superoxide dismutase 2 (SOD2) levels were increased in the tempol-treated group. In addition, the tempol-treated group showed reduced levels of glutathione-disulphide reductase (GSR), glutathione peroxidase 1 (GPx1) and catalase (CAT) in immunoblots. The tempol-treated group has also shown lower relative gene expression of SOD1, CAT and GPx than the non-treated group. Our data demonstrated that tempol treatment reduced oxidant parameters and increased anti-oxidant SOD2 levels in the DIA muscle of mdx mice, which may contribute to the normalization of the redox homeostasis of dystrophic muscles.
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme Luiz da Rocha
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Caroline Covatti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Elaine Cristina Leite Pereira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil.,Faculty of Ceilandia, University of Brasilia (UnB), Brasília, Brazil
| | - Renato Ferretti
- Department of Anatomy, Institute of Bioscience of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
17
|
Afjal MA, Goswami P, Ahmad S, Dabeer S, Akhter J, Salman M, Mangla A, Raisuddin S. Tempol (4-hydroxy tempo) protects mice from cisplatin-induced acute kidney injury via modulation of expression of aquaporins and kidney injury molecule-1. Drug Chem Toxicol 2020; 45:1355-1363. [PMID: 33078650 DOI: 10.1080/01480545.2020.1831011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tempol (4-hydroxy tempo), a pleiotropic antioxidant is reported to afford protection against cisplatin (CP)-induced nephrotoxicity. However, molecular mechanisms of action of tempol in improving the renal function in CP-induced nephrotoxicity are not fully understood. We investigated the attenuating effect of tempol against CP-induced alterations in kidney injury molecule-1 (KIM-1) and aquaporins (AQPs) in mice. Tempol (100 mg/kg, po) pretreatment with CP (20 mg/kg ip) showed restoration in renal function markers including electrolytes. CP treatment upregulated mRNA expression of KIM-1 and downregulated AQP and arginine vasopressin (AVP) expression which was attenuated by tempol. Immunoblotting analysis revealed that CP-induced alterations in KIM-1 and AQP expression were restored by tempol. Immunofluorocense study also showed restorative effect of tempol on the expression of AQP2 in CP-treated mice. In conclusion, this study provides experimental evidence that tempol resolved urinary concentration defect by the restoration of AQP, AVP and KIM-1 levels indicating a potential use of tempol in ameliorating the AKI in cancer patients under the treatment with CP.
Collapse
Affiliation(s)
- Mohd Amir Afjal
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Poonam Goswami
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Shahzad Ahmad
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Sadaf Dabeer
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Juheb Akhter
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Mohd Salman
- Molecular Neurobiology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Anuradha Mangla
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Sheikh Raisuddin
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| |
Collapse
|
18
|
Kumar S, Vinayak M. Quercetin Ameliorates CFA-Induced Chronic Inflammatory Hyperalgesia via Modulation of ROS-Mediated ERK1/2 Signaling and Inhibition of Spinal Glial Activation In Vivo. Neuromolecular Med 2020; 22:517-533. [DOI: 10.1007/s12017-020-08609-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 08/11/2020] [Indexed: 01/26/2023]
|
19
|
de Souza M, Baptista AAS, Valdiviezo MJJ, Justino L, Menck-Costa MF, Ferraz CR, da Gloria EM, Verri WA, Bracarense APFRL. Lactobacillus spp. reduces morphological changes and oxidative stress induced by deoxynivalenol on the intestine and liver of broilers. Toxicon 2020; 185:203-212. [PMID: 32687887 DOI: 10.1016/j.toxicon.2020.07.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/15/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
Abstract
The mycotoxin deoxynivalenol (DON) contaminates animal feed worldwide, frequently resulting in poor performance and economic losses. Data concerning the effects on poultry health or focusing on intestinal toxicity or the response to oxidative stress are scarce. Also, there is a need for strategies to mitigate the negative effects of DON. This study aimed to investigate the effects of Lactobacillus spp. treatments on the intestine, liver and kidney of poultry fed a DON-contaminated diet. To achieve this aim, histological, morphometrical and histochemical assays were performed. The oxidative stress response was also analyzed by the tests: reduced glutathione, ferric reducing ability, reducing of 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulphonic acid), nitro blue tetrazolium detection of superoxide anion, and thiobarbituric acid reactive substances. One-day-old broilers chickens (n 50) were submitted to the following treatments: control, DON (19.3 mg kg-1), viable Lactobacillus spp. + DON (VL + DON), heat-inactivated Lactobacillus spp. + DON (HIL + DON), Lactobacillus spp. culture supernatant + DON (LCS + DON). The animals received the contaminated diet for seven days. DON increased the intestinal and liver lesion score, while the Lactobacillus spp. treatments (LT) remained like the control. DON reduced the villi height and increased the crypt depths. The LT showed crypt depths similar to control, and higher villi: crypt ratio in duodenum and jejunum. In the ileum, the LT reduced the goblet cell count in relation to DON group. DON increased the number of intraepithelial lymphocytes (IEL) in jejunum and ileum, while the VL + DON treatment induced a significant decrease in IEL in comparison to DON. DON-diet induced an oxidative stress response in the intestine and liver, and also reduced the antioxidant capacity in these tissues, while LT treatments remained mostly similar to control. DON induced no change in redox balance in the kidney. The LT improved the intestinal health after DON acute exposure, reducing the oxidative stress damage mainly on jejunum and liver.
Collapse
Affiliation(s)
- Marielen de Souza
- Laboratory of Animal Pathology, Universidade Estadual de Londrina, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná, 86057-970, Brazil; Laboratory of Avian Medicine, Universidade Estadual de Londrina, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná, 86057-970, Brazil
| | - Ana Angelita S Baptista
- Laboratory of Avian Medicine, Universidade Estadual de Londrina, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná, 86057-970, Brazil
| | - Milton J J Valdiviezo
- Laboratory of Animal Pathology, Universidade Estadual de Londrina, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná, 86057-970, Brazil
| | - Larissa Justino
- Laboratory of Avian Medicine, Universidade Estadual de Londrina, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná, 86057-970, Brazil
| | - Maísa F Menck-Costa
- Laboratory of Avian Medicine, Universidade Estadual de Londrina, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná, 86057-970, Brazil
| | - Camila R Ferraz
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Universidade Estadual de Londrina, Londrina, Paraná, 86057-970, Brazil
| | | | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Universidade Estadual de Londrina, Londrina, Paraná, 86057-970, Brazil
| | - Ana Paula F R L Bracarense
- Laboratory of Animal Pathology, Universidade Estadual de Londrina, Campus Universitário, Rodovia Celso Garcia Cid, Km 380, Londrina, Paraná, 86057-970, Brazil.
| |
Collapse
|
20
|
Ye S, Xu P, Huang M, Chen X, Zeng S, Wang Q, Chen J, Li K, Gao W, Liu R, Liu J, Shao Y, Zhang H, Xu Y, Zhang Q, Zhong Z, Wei Z, Wang J, Hao B, Huang W, Liu Q. The heterocyclic compound Tempol inhibits the growth of cancer cells by interfering with glutamine metabolism. Cell Death Dis 2020; 11:312. [PMID: 32366855 PMCID: PMC7198543 DOI: 10.1038/s41419-020-2499-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
Tempol (4-hydroxy-2,2,6,6-Tetramethylpiperidine-1-oxyl, TPL), a nitroxide compound, inhibits proliferation and increases the vulnerability of cancer cells to apoptosis induced by cytotoxic agents. However, the molecular mechanism of TPL inhibiting cancer cell proliferation has not been fully understood. In this study, we evaluated the metabolic effect of TPL on cancer cells and explored its cancer therapeutic potential. Extracellular flow assays showed that TPL inhibited cellular basal and maximal oxygen consumption rates of mitochondrial. 13C metabolic flux analysis showed that TPL treatment had minimal effect on glycolysis. However, we found that TPL inhibits glutamine metabolism by interfering with the oxidative tricarboxylic acid cycle (TCA) process and reductive glutamine process. We found that the inhibitory effect of TPL on metabolism occurs mainly on the step from citrate to α-ketoglutarate or vice versa. We also found that activity of isocitrate dehydrogenase IDH1 and IDH2, the key enzymes in TCA, were inhibited by TPL treatment. In xenograft mouse model, TPL treatment reduced tumor growth by inhibiting cellular proliferation of xenograft tumors. Thus, we provided a mechanism of TPL inhibiting cancer cell proliferation by interfering with glutamine utilization that is important for survival and proliferation of cancer cells. The study may help the development of a therapeutic strategy of TPL combined with other anticancer medicines.
Collapse
Affiliation(s)
- Shuangyan Ye
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Pengfei Xu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Mengqiu Huang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xi Chen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Sisi Zeng
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qianli Wang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jianping Chen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Keyi Li
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenwen Gao
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ruiyuan Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Jingxian Liu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yihao Shao
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hui Zhang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yang Xu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qianbing Zhang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhuo Zhong
- Guangzhou Hospital of integrated Traditional and West Medicine, Guangzhou, China
| | - Zibo Wei
- Center for medical transformation, Shunde Hospital, Southern Medical University, Foshan, China
| | - Jiale Wang
- Center for medical transformation, Shunde Hospital, Southern Medical University, Foshan, China
| | - Bingtao Hao
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Wenhua Huang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China. .,Department of Human Anatomy, School of Basic Medical Sciences, Guangdong Medical University, Guangzhou, China.
| | - Qiuzhen Liu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China. .,Center for medical transformation, Shunde Hospital, Southern Medical University, Foshan, China.
| |
Collapse
|
21
|
Ye S, Zeng S, Huang M, Chen J, Chen X, Xu P, Wang Q, Gao W, Yang B, Hao B, Huang W, Liu Q. [Effect of the chemoprotectant tempol on anti-tumor activity of cisplatin]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:883-890. [PMID: 31511206 DOI: 10.12122/j.issn.1673-4254.2019.08.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the effect of the chemoprotectant tempol on the anti-tumor activity of cisplatin (DDP). METHODS The cellular toxicity of tempol in human colon cancer SW480 cells and mouse colon cancer CT26 cells were evaluated using MTT and cell counting kit-8 assays. CalcuSyn software analysis was used to determine the interaction between tempol and DDP in inhibition of the cell viability. A subcutaneous homograft mouse model of colon cancer was established. The mice were randomly divided into control group, tempol group, cisplatin group and tempol + DDP treatment group with intraperitoneal injections of the indicated agents. The tumor size, body weight and lifespan of the mice were measured, and HE staining was used to analyze the cytotoxic effect of the agents on the kidney and liver. Immunohistochemistry and Western blotting were performed to detect the expression of Bax and Bcl2 in the tumor tissue, and TUNEL staining was used to analyze the tumor cell apoptosis. The level of reactive oxygen species (ROS) in the tumor tissue was determined using flow cytometry. RESULTS Tempol showed inhibitory effects on the viability of SW480 and CT26 cells. CalcuSyn software analysis showed that tempol had a synergistic anti-tumor effect with DDP (CI < 1). In the homograft mouse model, tempol treatment alone did not produce obvious anti-tumor effect. HE staining showed that the combined use of tempol and DDP alleviated DDP-induced fibrogenesis in the kidneys, but tempol also reduced the anti-tumor activity of DDP. Compared with the mice treated with DDP alone, the mice treated with both tempol and DDP had a significantly larger tumor size (P < 0.01) and a shorter lifespan (P < 0.05). Tempol significantly reversed DDP-induced expression of Bax and Bcl2 in the tumor tissue and tumor cell apoptosis (P < 0.001), and obviously reduced the elevation of ROS level in the tumor tissue induced by DDP treatment (P < 0.05). CONCLUSIONS Tempol can attenuate the anti-tumor effect of DDP while reducing the side effects of DDP. Caution must be taken and the risks and benefits should be carefully weighed when considering the use of tempol as an anti-oxidant to reduce the toxicities of DDP.
Collapse
Affiliation(s)
- Shuangyan Ye
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Sisi Zeng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Mengqiu Huang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Jianping Chen
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Xi Chen
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Pengfei Xu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Qianli Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Wenwen Gao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Bingsheng Yang
- Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bingtao Hao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Wenhuan Huang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Department of Human Anatomy, School of Basic Medical Sciences, Guangdong Medical University, Zhanjiang 524003, China
| | - Qiuzhen Liu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China.,Center for Medical Transformation, Shunde Hospital, Southern Medical University, Foshan 528300, China
| |
Collapse
|
22
|
Direito R, Rocha J, Lima A, Gonçalves MM, Duarte MP, Mateus V, Sousa C, Fernandes A, Pinto R, Boavida Ferreira R, Sepodes B, Figueira ME. Reduction of Inflammation and Colon Injury by a Spearmint Phenolic Extract in Experimental Bowel Disease in Mice. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E65. [PMID: 31174376 PMCID: PMC6630206 DOI: 10.3390/medicines6020065] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
Background: Inflammatory Bowel Diseases (IBD) encompasses both Crohn's Disease and Ulcerative Colitis, known to be connected to an enlarged risk for developing colorectal cancer (CRC). Spearmint (Mentha spicata L.) is a Mediterranean plant used as an aromatic agent, and studies have mainly focused on the essential oil suggesting an anti-inflammatory activity. This work aimed to perform a preliminary screening of the in vivo anti-inflammatory effects of a spearmint phenolic extract in an acute inflammation model, in a chronic inflammation model of colitis, and also study the effects in vitro on a colon cancer model. Methods: Spearmint extract was administered to rats of a paw oedema model (induced by carrageenan) and to mice from a TNBS-induced colitis model in parallel with studies using HT-29 CRC cells. Results: Administration of the extract led to reduced paw inflammation, reduction of colon injury and inflammation, with attenuation of histological markers, and reduction of iNOS expression. It repressed the in vitro movement of HT-29 cells in a wound healing assay. Conclusions: These findings suggest that spearmint extract exhibits acute and chronic anti-inflammatory activity and is able to inhibit migration of cancer cells, suggesting a potential role in the supplementary therapy of IBD patients.
Collapse
Affiliation(s)
- Rosa Direito
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - João Rocha
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Ana Lima
- Disease & Stress Biology Group, LEAF, Instituto Superior de Agronomia, Universidade de Lisboa, 1349-017 Lisbon, Portugal.
| | - Maria Margarida Gonçalves
- Unidade de Biotecnologia Ambiental, Universidade Nova de Lisboa, Quinta da Torre, 2829-516 Monte da Caparica, Portugal.
| | - Maria Paula Duarte
- Unidade de Biotecnologia Ambiental (UBiA), Grupo de Disciplinas da Ecologia da Hidrosfera, Faculdade de Ciências e Tecnologia, FCT, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal.
| | - Vanessa Mateus
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
- H&TRC-Health and Technology Research Center, ESTeSL-Lisbon School of Health Technology, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal.
| | - Catarina Sousa
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Adelaide Fernandes
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Rui Pinto
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
- Joaquim Chaves Saúde, Dr Joaquim Chaves Lab Analises Clínicas, 1495-068 Miraflores-Algés, Portugal.
| | - Ricardo Boavida Ferreira
- Disease & Stress Biology Group, LEAF, Instituto Superior de Agronomia, Universidade de Lisboa, 1349-017 Lisbon, Portugal.
| | - Bruno Sepodes
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Maria-Eduardo Figueira
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| |
Collapse
|
23
|
Flavin Oxidase-Induced ROS Generation Modulates PKC Biphasic Effect of Resveratrol on Endothelial Cell Survival. Biomolecules 2019; 9:biom9060209. [PMID: 31151226 PMCID: PMC6628153 DOI: 10.3390/biom9060209] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/22/2019] [Accepted: 05/25/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Dietary intake of natural antioxidants is thought to impart protection against oxidative-associated cardiovascular diseases. Despite many in vivo studies and clinical trials, this issue has not been conclusively resolved. Resveratrol (RES) is one of the most extensively studied dietary polyphenolic antioxidants. Paradoxically, we have previously demonstrated that high RES concentrations exert a pro-oxidant effect eventually elevating ROS levels leading to cell death. Here, we further elucidate the molecular determinants underpinning RES-induced oxidative cell death. Methods: Using human umbilical vein endothelial cells (HUVECs), the effect of increasing concentrations of RES on DNA synthesis and apoptosis was studied. In addition, mRNA and protein levels of cell survival or apoptosis genes, as well as protein kinase C (PKC) activity were determined. Results: While high concentrations of RES reduce PKC activity, inhibit DNA synthesis and induce apoptosis, low RES concentrations elicit an opposite effect. This biphasic concentration-dependent effect (BCDE) of RES on PKC activity is mirrored at the molecular level. Indeed, high RES concentrations upregulate the proapoptotic Bax, while downregulating the antiapoptotic Bcl-2, at both mRNA and protein levels. Similarly, high RES concentrations downregulate the cell cycle progression genes, c-myc, ornithine decarboxylase (ODC) and cyclin D1 protein levels, while low RES concentrations display an increasing trend. The BCDE of RES on PKC activity is abrogated by the ROS scavenger Tempol, indicating that this enzyme acts downstream of the RES-elicited ROS signaling. The RES-induced BCDE on HUVEC cell cycle machinery was also blunted by the flavin inhibitor diphenyleneiodonium (DPI), implicating flavin oxidase-generated ROS as the mechanistic link in the cellular response to different RES concentrations. Finally, PKC inhibition abrogates the BCDE elicited by RES on both cell cycle progression and pro-apoptotic gene expression in HUVECs, mechanistically implicating PKC in the cellular response to different RES concentrations. Conclusions: Our results provide new molecular insight into the impact of RES on endothelial function/dysfunction, further confirming that obtaining an optimal benefit of RES is concentration-dependent. Importantly, the BCDE of RES could explain why other studies failed to establish the cardio-protective effects mediated by natural antioxidants, thus providing a guide for future investigation looking at cardio-protection by natural antioxidants.
Collapse
|
24
|
Hermes TDA, Mâncio RD, Macedo AB, Mizobuti DS, da Rocha GL, Cagnon VHA, Minatel E. Tempol treatment shows phenotype improvement in mdx mice. PLoS One 2019; 14:e0215590. [PMID: 31009514 PMCID: PMC6476507 DOI: 10.1371/journal.pone.0215590] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/04/2019] [Indexed: 01/04/2023] Open
Abstract
Considering potential Tempol effects on mdx muscle fibers, in this study we evaluated its effects on relevant dystrophic phenotypic characteristics, such as muscle degeneration, inflammatory process and angiogenesis, which as yet have not been investigated. Mdx mice were randomly assigned into three groups: mdxS, the control group receiving intraperitoneal (i.p.) injections of saline solution (100μL); mdxP, positive control group receiving prednisolone (1mg/kg) by oral gavage; and mdxT, treated group receiving i.p. injections of tempol (100 mg/kg). C57BL/10 mice were also used as controls. Tempol treatment promoted gain in muscle strength and reduced myonecrosis and inflammatory response in the dystrophic diaphragm (DIA) and biceps brachii (BB) muscles. No evidence of Tempol's beneficial performance on angiogenesis in DIA and BB mdx muscles was found. The findings presented here show that Tempol treatment improves dystrophic phenotype, supporting its use as a potential therapeutic strategy in DMD.
Collapse
MESH Headings
- Animals
- Antioxidants/administration & dosage
- Antioxidants/pharmacology
- Cyclic N-Oxides/administration & dosage
- Cyclic N-Oxides/pharmacology
- Diaphragm/metabolism
- Diaphragm/physiopathology
- Disease Models, Animal
- Humans
- Injections, Intraperitoneal
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/physiology
- Muscle Strength/drug effects
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Muscular Dystrophies/genetics
- Muscular Dystrophies/pathology
- Muscular Dystrophies/physiopathology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/physiopathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Phenotype
- Spin Labels
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Rafael Dias Mâncio
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Aline Barbosa Macedo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Guilherme Luiz da Rocha
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Valéria Helena Alves Cagnon
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
25
|
Repurposing of the Nootropic Drug Vinpocetine as an Analgesic and Anti-Inflammatory Agent: Evidence in a Mouse Model of Superoxide Anion-Triggered Inflammation. Mediators Inflamm 2019; 2019:6481812. [PMID: 31049025 PMCID: PMC6462340 DOI: 10.1155/2019/6481812] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/27/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Clinically active drugs for the treatment of acute pain have their prescription limited due to the significant side effects they induce. An increase in reactive oxygen species (ROS) has been linked to several conditions, including inflammation and pain processing. Therefore, new or repurposed drugs with the ability of reducing ROS-triggered responses are promising candidates for analgesic drugs. Vinpocetine is a clinically used nootropic drug with antioxidant, anti-inflammatory, and analgesic properties. However, the effects of vinpocetine have not been investigated in a model with a direct relationship between ROS, inflammation, and pain. Based on that, we aimed to investigate the effects of vinpocetine in a model of superoxide anion-induced pain and inflammation using potassium superoxide (KO2) as a superoxide anion donor to trigger inflammation and pain. In the KO2 model, vinpocetine dose-dependently reduced pain-like behaviors (spontaneous pain and hyperalgesia), paw edema, and neutrophil and mononuclear cell recruitment to the paw skin (assessed by H&E staining, fluorescence, and enzymatic assays) and to the peritoneal cavity. Vinpocetine also restored tissue endogenous antioxidant ability and Nrf2 and Ho-1 mRNA expression and reduced superoxide anion production and gp91phox mRNA expression. We also observed the inhibition of IκBα degradation by vinpocetine, which demonstrates a reduction in the activation of NF-κB explaining the diminished production of IL-33, IL-1β, and TNF-α. Collectively, our data show that vinpocetine alleviates pain and inflammation induced by KO2, which is a mouse model with a direct role of ROS in triggering pain and other inflammatory phenomena. Thus, the results suggest the repurposing of vinpocetine as an anti-inflammatory and analgesic drug.
Collapse
|
26
|
Bussmann AJC, Borghi SM, Zaninelli TH, Dos Santos TS, Guazelli CFS, Fattori V, Domiciano TP, Pinho-Ribeiro FA, Ruiz-Miyazawa KW, Casella AMB, Vignoli JA, Camilios-Neto D, Casagrande R, Verri WA. The citrus flavanone naringenin attenuates zymosan-induced mouse joint inflammation: induction of Nrf2 expression in recruited CD45 + hematopoietic cells. Inflammopharmacology 2019; 27:1229-1242. [PMID: 30612217 DOI: 10.1007/s10787-018-00561-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/31/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Naringenin is a biologically active analgesic, anti-inflammatory, and antioxidant flavonoid. Naringenin targets in inflammation-induced articular pain remain poorly explored. METHODS The present study investigated the cellular and molecular mechanisms involved in the analgesic/anti-inflammatory effects of naringenin in zymosan-induced arthritis. Mice were pre-treated orally with naringenin (16.7-150 mg/kg), followed by intra-articular injection of zymosan. Articular mechanical hyperalgesia and oedema, leucocyte recruitment to synovial cavity, histopathology, expression/production of pro- and anti-inflammatory mediators and NFκB activation, inflammasome component expression, and oxidative stress were evaluated. RESULTS Naringenin inhibited articular pain and oedema in a dose-dependent manner. The dose of 50 mg/kg inhibited leucocyte recruitment, histopathological alterations, NFκB activation, and NFκB-dependent pro-inflammatory cytokines (TNF-α, IL-1β, and IL-33), and preproET-1 mRNA expression, but increased anti-inflammatory IL-10. Naringenin also inhibited inflammasome upregulation (reduced Nlrp3, ASC, caspase-1, and pro-IL-1β mRNA expression) and oxidative stress (reduced gp91phox mRNA expression and superoxide anion production, increased GSH levels, induced Nrf2 protein in CD45+ hematopoietic recruited cells, and induced Nrf2 and HO-1 mRNA expression). CONCLUSIONS Naringenin presents analgesic and anti-inflammatory effects in zymosan-induced arthritis by targeting its main physiopathological mechanisms. These data highlight this flavonoid as an interesting therapeutic compound to treat joint inflammation, deserving additional pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Allan J C Bussmann
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Sergio M Borghi
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Tiago H Zaninelli
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Telma S Dos Santos
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Carla F S Guazelli
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Victor Fattori
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Talita P Domiciano
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Felipe A Pinho-Ribeiro
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Kenji W Ruiz-Miyazawa
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil
| | - Antonio M B Casella
- Department of Clinical Medicine, Health Science Center, Londrina State University, University Hospital, 86039-440, Londrina, Paraná State, Brazil
| | - Josiane A Vignoli
- Department of Biochemistry and Biotechnology, Exact Sciences Center, Londrina State University, Londrina, 86057-970, Brazil
| | - Doumit Camilios-Neto
- Department of Biochemistry and Biotechnology, Exact Sciences Center, Londrina State University, Londrina, 86057-970, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Health Science Center, Londrina State University, University Hospital, Londrina, Paraná State, 86039-440, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Biological Science Center, Londrina State University, Rod. Celso Garcia Cid, PR 445, Km 380, Londrina, Paraná State, 86051-990, Brazil.
| |
Collapse
|
27
|
Reactive Oxygen Species from NADPH Oxidase and Mitochondria Participate in the Proliferation of Aortic Smooth Muscle Cells from a Model of Metabolic Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5835072. [PMID: 30671170 PMCID: PMC6323422 DOI: 10.1155/2018/5835072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/27/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
In metabolic diseases, the increased reactive oxygen species (ROS) represents one of the pathogenic mechanisms for vascular disease probably by promoting vascular smooth muscle cell (SMC) proliferation that contributes to the development of arterial remodeling and stenosis, hypertension, and atherosclerosis. Therefore, this work was undertaken to evaluate the participation of ROS from NADPH oxidase and mitochondria in the proliferation of SMCs from the aorta in a model of metabolic syndrome induced by sucrose feeding in rats. After 24 weeks, sucrose-fed (SF) rats develop hypertension, intra-abdominal obesity, hyperinsulinemia, and hyperleptinemia. In addition SMCs from SF rats had a higher growth rate and produce more ROS than control cells. The treatment of SMCs with DPI and apocynin to inhibit NADPH oxidase and with tempol to scavenge superoxide anion significantly blocked the proliferation of both SF and control cells suggesting the participation of NADPH oxidase as a source of superoxide anion. MitoTEMPO, which targets mitochondria within the cell, also significantly inhibited the proliferation of SMCs having a greater effect on cells from SF than from the control aorta. The higher rate of cell growth from the SF aorta is supported by the increased content of cyclophilin A and CD147, proteins involved in the mechanism of cell proliferation. In addition, caldesmon, α-actin, and phosphorylated myosin light chain, contractile phenotype proteins, were found significantly lower in SF cells in no confluent state and increased in confluent state but without difference between both cell types. Our results suggest that ROS from NADPH oxidase and mitochondria significantly participate in the difference found in the rate of cell growth between SF and control cells.
Collapse
|
28
|
Li L, Guo J, Wang Y, Xiong X, Tao H, Li J, Jia Y, Hu H, Zhang J. A Broad-Spectrum ROS-Eliminating Material for Prevention of Inflammation and Drug-Induced Organ Toxicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800781. [PMID: 30356945 PMCID: PMC6193162 DOI: 10.1002/advs.201800781] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/22/2018] [Indexed: 05/12/2023]
Abstract
Despite the great potential of numerous antioxidants for pharmacotherapy of diseases associated with inflammation and oxidative stress, many challenges remain for their clinical translation. Herein, a superoxidase dismutase/catalase-mimetic material based on Tempol and phenylboronic acid pinacol ester simultaneously conjugated β-cyclodextrin (abbreviated as TPCD), which is capable of eliminating a broad spectrum of reactive oxygen species (ROS), is reported. TPCD can be easily synthesized by sequentially conjugating two functional moieties onto a β-cyclodextrin scaffold. The thus developed pharmacologically active material may be easily produced into antioxidant and anti-inflammatory nanoparticles, with tunable size. TPCD nanoparticles (TPCD NP) effectively protect macrophages from oxidative stress-induced apoptosis in vitro. Consistently, TPCD NP shows superior efficacies in three murine models of inflammatory diseases, with respect to attenuating inflammatory responses and mitigating oxidative stress. TPCD NP can also protect mice from drug-induced organ toxicity. Besides the passive targeting effect, the broad spectrum ROS-scavenging capability contributes to the therapeutic benefits of TPCD NP. Importantly, in vitro and in vivo preliminary experiments demonstrate the good safety profile of TPCD NP. Consequently, TPCD in its native and nanoparticle forms can be further developed as efficacious and safe therapies for treatment of inflammation and oxidative stress-associated diseases.
Collapse
Affiliation(s)
- Lanlan Li
- Department of PharmaceuticsCollege of PharmacyThird Military Medical UniversityChongqing400038China
| | - Jiawei Guo
- Department of PharmaceuticsCollege of PharmacyThird Military Medical UniversityChongqing400038China
| | - Yuquan Wang
- Department of CardiologySouthwest HospitalThird Military Medical UniversityChongqing400038China
- Department of CardiologyAffiliated Hospital of North Sichuan Medical CollegeNanchong637000Sichuan ProvinceChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Hui Tao
- Department of PharmaceuticsCollege of PharmacyThird Military Medical UniversityChongqing400038China
| | - Jin Li
- Department of PharmaceuticsCollege of PharmacyThird Military Medical UniversityChongqing400038China
| | - Yi Jia
- Department of PharmaceuticsCollege of PharmacyThird Military Medical UniversityChongqing400038China
| | - Houyuan Hu
- Department of CardiologySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Jianxiang Zhang
- Department of PharmaceuticsCollege of PharmacyThird Military Medical UniversityChongqing400038China
| |
Collapse
|
29
|
Wert KJ, Velez G, Cross MR, Wagner BA, Teoh-Fitzgerald ML, Buettner GR, McAnany JJ, Olivier A, Tsang SH, Harper MM, Domann FE, Bassuk AG, Mahajan VB. Extracellular superoxide dismutase (SOD3) regulates oxidative stress at the vitreoretinal interface. Free Radic Biol Med 2018; 124:408-419. [PMID: 29940351 PMCID: PMC6233711 DOI: 10.1016/j.freeradbiomed.2018.06.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
Oxidative stress is a pathogenic feature in vitreoretinal disease. However, the ability of the inner retina to manage metabolic waste and oxidative stress is unknown. Proteomic analysis of antioxidants in the human vitreous, the extracellular matrix opposing the inner retina, identified superoxide dismutase-3 (SOD3) that localized to a unique matrix structure in the vitreous base and cortex. To determine the role of SOD3, Sod3-/- mice underwent histological and clinical phenotyping. Although the eyes were structurally normal, at the vitreoretinal interface Sod3-/- mice demonstrated higher levels of 3-nitrotyrosine, a key marker of oxidative stress. Pattern electroretinography also showed physiological signaling abnormalities within the inner retina. Vitreous biopsies and epiretinal membranes collected from patients with diabetic vitreoretinopathy (DVR) and a mouse model of DVR showed significantly higher levels of nitrates and/or 3-nitrotyrosine oxidative stress biomarkers suggestive of SOD3 dysfunction. This study analyzes the molecular pathways that regulate oxidative stress in human vitreous substructures. The absence or dysregulation of the SOD3 antioxidant at the vitreous base and cortex results in increased oxidative stress and tissue damage to the inner retina, which may underlie DVR pathogenesis and other vitreoretinal diseases.
Collapse
Affiliation(s)
- Katherine J Wert
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, United States; Omics Laboratory, Stanford University, Palo Alto, CA, United States
| | - Gabriel Velez
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, United States; Omics Laboratory, Stanford University, Palo Alto, CA, United States
| | - Madeline R Cross
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Brett A Wagner
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Melissa L Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Garry R Buettner
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - J Jason McAnany
- Department of Ophthalmology, University of Illinois at Chicago, Chicago, IL, United States
| | - Alicia Olivier
- Division of Comparative Pathology, Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Stephen H Tsang
- Bernard and Shirlee Brown Glaucoma Laboratory and Barbara & Donald Jonas Laboratory of Regenerative Medicine, Columbia University, New York, NY, United States; Edward S. Harkness Eye Institute, Columbia University, New York, NY, United States; Departments of Ophthalmology, Pathology & Cell Biology, and Institute of Human Nutrition, Columbia University, New York, NY, United States
| | - Matthew M Harper
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States; Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA, United States; Veterans Affairs Center for the Prevention and Treatment of Visual Loss, Iowa City, IA, United States
| | - Frederick E Domann
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Vinit B Mahajan
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, United States; Omics Laboratory, Stanford University, Palo Alto, CA, United States; Palo Alto Veterans Administration, Palo Alto, CA, United States.
| |
Collapse
|
30
|
Analgesic Effect of Methane Rich Saline in a Rat Model of Chronic Inflammatory Pain. Neurochem Res 2018; 43:869-877. [PMID: 29411262 DOI: 10.1007/s11064-018-2490-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/08/2018] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
Abstract
How oxidative stress contributes to neuro-inflammation and chronic pain is documented, and methane is reported to protect against ischemia-reperfusion injury in the nervous system via anti-inflammatory and antioxidant properties. We studied whether methane in the form of methane rich saline (MS) has analgesic effects in a monoarthritis (MA) rat model of chronic inflammatory pain. Single and repeated injections of MS (i.p.) reduced MA-induced mechanical allodynia and multiple methane treatments blocked activation of glial cells, decreased IL-1β and TNF-α production and MMP-2 activity, and upregulated IL-10 expression in the spinal cord on day 10 post-MA. Furthermore, MS reduced infiltrating T cells and expression of IFN-γ and suppressed MA-induced oxidative stress (MDA and 8-OHDG), and increased superoxide dismutase and catalase activity. Thus, MS may offer anti-inflammatory and antioxidant effects to reduce chronic inflammatory pain.
Collapse
|
31
|
Bonetta R. Potential Therapeutic Applications of MnSODs and SOD-Mimetics. Chemistry 2017; 24:5032-5041. [DOI: 10.1002/chem.201704561] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Rosalin Bonetta
- Centre of Molecular Medicine and Biobanking; University of Malta; Msida MSD2080 Malta
| |
Collapse
|