1
|
Hao J, Ye Y, Zhang G, Shen H, Li J, Chen G. Mechanisms of nitric oxide in spinal cord injury. Med Gas Res 2024; 14:192-200. [PMID: 39073327 DOI: 10.4103/mgr.medgasres-d-23-00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/04/2023] [Indexed: 07/30/2024] Open
Abstract
Spinal cord injury (SCI) is a primary lesion of the spinal cord that results from external forces or diseases, accompanied by a cascade of secondary events. Nitric oxide, an endogenous gas that functions as a signaling molecule in the human body, plays a crucial role in vasodilation of smooth muscles, regulation of blood flow and pressure, and inflammatory response. This article provides a comprehensive overview of the involvement of nitric oxide in SCI and highlights recent advances in basic research on pharmacological agents that inhibit nitric oxide elevation after SCI, offering valuable insights for future therapeutic interventions targeting SCI.
Collapse
Affiliation(s)
- Jiahui Hao
- Brain and Nerve Research Laboratory, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
2
|
Firdous SM, Khan SA, Maity A. Oxidative stress-mediated neuroinflammation in Alzheimer's disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8189-8209. [PMID: 38832985 DOI: 10.1007/s00210-024-03188-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/24/2024] [Indexed: 06/06/2024]
Abstract
Reactive oxygen species (ROS) are metabolic by-products that constitute an indispensable component of physiological processes, albeit their heightened presence may proffer substantial perils to biological entities. Such a proliferation gives rise to a gradual escalation of oxidative stress within the organism, thereby compromising mitochondrial functionality and inflicting harm upon various bodily systems, with a particular predilection for the central nervous system. In its nascent stages, it is plausible that inflammation has been a facilitator in the progression of the malady. The precise role of inflammation in Alzheimer's disease (AD) remains somewhat enigmatic, although it is conceivable that activated microglia and astrocytes might be implicated in the removal of amyloid-β (Aβ) deposits. Nonetheless, prolonged microglial activation is associated with Tau phosphorylation and Aβ aggregation. Research studies have indicated that AD brains upregulate complementary molecules, inflammatory cytokines, acute phase reacting agents, and other inflammatory mediators that may cause neurodegeneration. In this review, oxidative damage products will be discussed as potential peripheral biomarkers for AD and its early stages. The disordered excretion of pro-inflammatory cytokines, chemokines, oxygen, and nitrogen-reactive species, along with the stimulation of the complement system by glial cells, has the potential to disrupt the functionality of neuronal termini. This perturbation, in turn, culminates in compromised synaptic function, a phenomenon empirically linked to the manifestation of cognitive impairments. The management of neurodegenerative conditions in the context of dementia necessitates therapeutic interventions that specifically target the excessive production of inflammatory and oxidative agents. Furthermore, we shall deliberate upon the function of microglia and oxidative injury in the etiology of AD and the ensuing neurodegenerative processes.
Collapse
Affiliation(s)
- Sayed Mohammed Firdous
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, 711316, West Bengal, India.
| | - Sahabaj Ali Khan
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, 711316, West Bengal, India
| | - Amritangshu Maity
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, 711316, West Bengal, India
| |
Collapse
|
3
|
Janarathanam VA, Issac PK, Pan I, Kamaraj N, Ansar S, Kumar YA, Guru A. Investigating antioxidant effects of hamamelitannin-conjugated zinc oxide nanoparticles on oxidative stress-Induced neurotoxicity in zebrafish larvae model. Mol Biol Rep 2024; 51:1087. [PMID: 39436450 DOI: 10.1007/s11033-024-09998-1] [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: 08/06/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND An excessive amount of reactive oxygen species triggers oxidative stress, leading to an imbalance in cellular homeostasis. Antioxidant therapy is an effective tool for lowering the oxidative stress and associated ailments. Recently, green nano-based drug formulations have demonstrated promising antioxidant activity and neutralizing oxidative stress. In this study, a tannin molecule Hamamelitannin (HAM), was utilized to synthesize zinc oxide nanoparticles HAM-ZnO NPs, to mitigate oxidative stress and associated ailments . METHODOLOGY The HAM-ZnO NPs were synthesized and characterized by XRD, SEM, and FTIR. The antioxidant potentials of HAM-ZnO NPs were analyzed by in vitro antioxidant assays. Zebrafish embryos and larvae were used as in-vivo models to assess the toxicity and antioxidant protective mechanism. Hydrogen peroxide (1mM) was employed to induce oxidative stress and treated with HAM-ZnO NPs to study the cognitive impairment and antioxidant enzyme levels. Levels of reactive oxygen species and cell death due to oxidative stress induction were studied by 2',7'-dichlorodihydrofluorescein diacetate and Acridine orange staining methods. Additionally, expression of Antioxidant genes such as SOD, CAT, GPx, and GSR were studied. . RESULTS HAM-ZnO NPs exhibited a spherical morphology and size ranges between 48 and 53 nm. In vitro antioxidant studies revealed the antioxidant properties of HAM-ZnO NPs. Furthermore, in vivo studies indicated that HAM-ZnO NPs don't possess any cytotoxic effects in zebrafish larvae at concentrations between (5-25 µg/ml), The study also observed that HAM-ZnO NPs significantly reduced Hydrogen Peroxide-induced stress and increased antioxidant activity in zebrafish larvae. Also, the antioxidant gene expression was upregulated in the HAM-ZnO NPs zebrafish larvae. CONCLUSION Findings in this study showed that HAM-ZnO NPs might be a potential intervention for diseases linked to oxidative stress.
Collapse
Affiliation(s)
- Vishnu Adith Janarathanam
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602 105, Tamil Nadu, India
| | - Praveen Kumar Issac
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602 105, Tamil Nadu, India.
| | - Ieshita Pan
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602 105, Tamil Nadu, India
| | - Nagalakshmi Kamaraj
- Department of Biotechnology, Karpaga Vinayaga College of Engineering and Technology, Chinna Kolambakkam, Chengalpattu District, Padalam, India
| | - Sabah Ansar
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Yedluri Anil Kumar
- Department of Chemistry, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Ajay Guru
- Department of Cariology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India
| |
Collapse
|
4
|
Mazuryk O, Gurgul I, Oszajca M, Polaczek J, Kieca K, Bieszczad-Żak E, Martyka T, Stochel G. Nitric Oxide Signaling and Sensing in Age-Related Diseases. Antioxidants (Basel) 2024; 13:1213. [PMID: 39456466 PMCID: PMC11504650 DOI: 10.3390/antiox13101213] [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: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Nitric oxide (NO) is a key signaling molecule involved in numerous physiological and pathological processes within the human body. This review specifically examines the involvement of NO in age-related diseases, focusing on the cardiovascular, nervous, and immune systems. The discussion delves into the mechanisms of NO signaling in these diseases, emphasizing the post-translational modifications of involved proteins, such as S-nitrosation and nitration. The review also covers the dual nature of NO, highlighting both its protective and harmful effects, determined by concentration, location, and timing. Additionally, potential therapies that modulate NO signaling, including the use of NO donors and nitric oxide synthases (NOSs) inhibitors in the treatment of cardiovascular, neurodegenerative, and oncological diseases, are analyzed. Particular attention is paid to the methods for the determination of NO and its derivatives in the context of illness diagnosis and monitoring. The review underscores the complexity and dual role of NO in maintaining cellular balance and suggests areas for future research in developing new therapeutic strategies.
Collapse
Affiliation(s)
- Olga Mazuryk
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Ilona Gurgul
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Maria Oszajca
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Justyna Polaczek
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Konrad Kieca
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Ewelina Bieszczad-Żak
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Tobiasz Martyka
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Grażyna Stochel
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| |
Collapse
|
5
|
Bazbaz W, Kartawy M, Hamoudi W, Ojha SK, Khaliulin I, Amal H. The Role of Thioredoxin System in Shank3 Mouse Model of Autism. J Mol Neurosci 2024; 74:90. [PMID: 39347996 PMCID: PMC11457715 DOI: 10.1007/s12031-024-02270-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by difficulties in social interaction and communication, repetitive behaviors, and restricted interests. Unfortunately, the underlying molecular mechanism behind ASD remains unknown. It has been reported that oxidative and nitrosative stress are strongly linked to ASD. We have recently found that nitric oxide (NO•) and its products play an important role in this disorder. One of the key proteins associated with NO• is thioredoxin (Trx). We hypothesize that the Trx system is altered in the Shank3 KO mouse model of autism, which may lead to a decreased activity of the nuclear factor erythroid 2-related factor 2 (Nrf2), resulting in oxidative stress, and thus, contributing to ASD-related phenotypes. To test this hypothesis, we conducted in vivo behavioral studies and used primary cortical neurons derived from the Shank3 KO mice and human SH-SY5Y cells with SHANK3 mutation. We showed significant changes in the levels and activity of Trx redox proteins in the Shank3 KO mice. A Trx1 inhibitor PX-12 decreased Trx1 and Nrf2 expression in wild-type mice, causing abnormal alterations in the levels of synaptic proteins and neurotransmission markers, and an elevation of nitrosative stress. Trx inhibition resulted in an ASD-like behavioral phenotype, similar to that of Shank3 KO mice. Taken together, our findings confirm the strong link between the Trx system and ASD pathology, including the increased oxidative/nitrosative stress, and synaptic and behavioral deficits. The results of this study may pave the way for identifying novel drug targets for ASD.
Collapse
Affiliation(s)
- Wisam Bazbaz
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
6
|
Pino MTL, Rocca MV, Acosta LH, Cabilla JP. Challenging the Norm: The Unrecognized Impact of Soluble Guanylyl Cyclase Subunits in Cancer. Int J Mol Sci 2024; 25:10053. [PMID: 39337539 PMCID: PMC11432225 DOI: 10.3390/ijms251810053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Since the discovery of nitric oxide (NO), a long journey has led us to the present, during which much knowledge has been gained about its pathway members and their roles in physiological and various pathophysiological conditions. Soluble guanylyl cyclase (sGC), the main NO receptor composed of the sGCα1 and sGCβ1 subunits, has been one of the central figures in this narrative. However, the sGCα1 and sGCβ1 subunits remained obscured by the focus on sGC's enzymatic activity for many years. In this review, we restore the significance of the sGCα1 and sGCβ1 subunits by compiling and analyzing available but previously overlooked information regarding their roles beyond enzymatic activity. We delve into the basics of sGC expression regulation, from its transcriptional regulation to its interaction with proteins, placing particular emphasis on evidence thus far demonstrating the actions of each sGC subunit in different tumor models. Exploring the roles of sGC subunits in cancer offers a valuable opportunity to enhance our understanding of tumor biology and discover new therapeutic avenues.
Collapse
Affiliation(s)
- María Teresa L Pino
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| | - María Victoria Rocca
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| | - Lucas H Acosta
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| | - Jimena P Cabilla
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, CONICET-Universidad Abierta Interamericana, Buenos Aires C1270AAH, Argentina
| |
Collapse
|
7
|
Al-Madhagi H, Masoud A. Limitations and Challenges of Antioxidant Therapy. Phytother Res 2024. [PMID: 39260385 DOI: 10.1002/ptr.8335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
Our bodies are constantly exposed to or producing free radicals nearly on a daily basis. These highly reactive molecules are generated through a variety of internal and external processes and pathways within the body. If these free radicals are not neutralized by antioxidants, they can lead to a state of oxidative stress, which has been linked to a wide range of severe and debilitating disorders affecting various systems in the human body. This involves neurodegenerative diseases, diabetes, atherosclerosis, fatty liver, inflammation, and aging. Thankfully, the human body is armed with a repertoire of powerful antioxidants with different natures and modes of action. The recent decades witnessed the publication of enormous papers proving antioxidant activity of a novel synthesized compound, plant extract, or a purified drug in vitro, in vivo, and even on human beings. However, the efficacy of antioxidant therapies in clinical trials, including selenium, vitamin C, vitamin E, and vitamin A, has been notably inconsistent. This inconsistency can be primarily ascribed to different factors related to the nature of free radical generation, purpose and the specific type of therapy employed, and the intricate oxidative stress connected network, among others. Collectively, these factors will be explored in this review article to decipher the observed shortcomings in the application of antioxidant therapies within clinical settings.
Collapse
Affiliation(s)
| | - Anwar Masoud
- Biochemical Technology Program, Dhamar University, Dhamar, Yemen
| |
Collapse
|
8
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2024:10.1038/s41380-024-02725-z. [PMID: 39223276 DOI: 10.1038/s41380-024-02725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
9
|
Yang J, Qin G, Liu Z, Zhang H, Du X, Ren J, Qu X. A Nanozyme-Boosted MOF-CRISPR Platform for Treatment of Alzheimer's Disease. NANO LETTERS 2024; 24:9906-9915. [PMID: 39087644 DOI: 10.1021/acs.nanolett.4c02272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Rectifying the aberrant microenvironment of a disease through maintenance of redox homeostasis has emerged as a promising perspective with significant therapeutic potential for Alzheimer's disease (AD). Herein, we design and construct a novel nanozyme-boosted MOF-CRISPR platform (CMOPKP), which can maintain redox homeostasis and rescue the impaired microenvironment of AD. By modifying the targeted peptides KLVFFAED, CMOPKP can traverse the blood-brain barrier and deliver the CRISPR activation system for precise activation of the Nrf2 signaling pathway and downstream redox proteins in regions characterized by oxidative stress, thereby reinstating neuronal antioxidant capacity and preserving redox homeostasis. Furthermore, cerium dioxide possessing catalase enzyme-like activity can synergistically alleviate oxidative stress. Further in vivo studies demonstrate that CMOPKP can effectively alleviate cognitive impairment in 3xTg-AD mouse models. Therefore, our design presents an effective way for regulating redox homeostasis in AD, which shows promise as a therapeutic strategy for mitigating oxidative stress in AD.
Collapse
Affiliation(s)
- Jie Yang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| | - Geng Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| | - Zhenqi Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| | - Haochen Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| | - Xiubo Du
- College of Life Sciences and Oceanography, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
| |
Collapse
|
10
|
Karkoszka N, Gibula-Tarlowska E, Kotlinska J, Bielenica A, Gawel K, Kedzierska E. Selenium Intake and Postnatal Depression-A Short Review. Nutrients 2024; 16:1926. [PMID: 38931280 PMCID: PMC11206929 DOI: 10.3390/nu16121926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Postnatal depression is a common and severe complication of childbirth. It is an important public health problem with significant implications for both mothers and children. The exact mechanisms underlying and the factors influencing the occurrence of postnatal depression remain unclear. The literature suggests that certain dietary deficiencies during pregnancy and the postnatal period may contribute to a greater risk of maternal depression. This review focuses on the role of selenium in postnatal depression. It collects evidence from published interventional and observational studies investigating the relationship between selenium intake during the antenatal and postnatal periods and the mental status of postpartum women and summarises information about biological mechanisms that may underlie the association between selenium status and postnatal depression. The review includes studies identified through electronic searches of Medline (via PubMed) and Google Scholar databases until December 2023. Despite the small number of relevant studies and their potential methodological limitations, the findings suggest that optimizing selenium status may support the prevention and treatment of postnatal depression. Further longitudinal and interventional studies are necessary to confirm the clinical significance of these effects.
Collapse
Affiliation(s)
- Natalia Karkoszka
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4a Chodźki Street, 20-400 Lublin, Poland; (E.G.-T.); (J.K.); (E.K.)
| | - Ewa Gibula-Tarlowska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4a Chodźki Street, 20-400 Lublin, Poland; (E.G.-T.); (J.K.); (E.K.)
| | - Jolanta Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4a Chodźki Street, 20-400 Lublin, Poland; (E.G.-T.); (J.K.); (E.K.)
| | - Anna Bielenica
- Department of Biochemistry, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
| | - Kinga Gawel
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, 8b Jaczewskiego Street, 20-090 Lublin, Poland;
| | - Ewa Kedzierska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4a Chodźki Street, 20-400 Lublin, Poland; (E.G.-T.); (J.K.); (E.K.)
| |
Collapse
|
11
|
Pidany F, Kroustkova J, Jenco J, Breiterova KH, Muckova L, Novakova L, Kunes J, Fibigar J, Kucera T, Novak M, Sorf A, Hrabinova M, Pulkrabkova L, Janousek J, Soukup O, Jun D, Korabecny J, Cahlikova L. Carltonine-derived compounds for targeted butyrylcholinesterase inhibition. RSC Med Chem 2024; 15:1601-1625. [PMID: 38784455 PMCID: PMC11110763 DOI: 10.1039/d4md00060a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/16/2024] [Indexed: 05/25/2024] Open
Abstract
The investigation into human butyrylcholinesterase (hBChE) inhibitors as therapeutic agents for Alzheimer's disease (AD) holds significant promise, addressing both symptomatic relief and disease progression. In the pursuit of novel drug candidates with a selective BChE inhibition pattern, we focused on naturally occurring template structures, specifically Amaryllidaceae alkaloids of the carltonine-type. Herein, we explored a series of compounds implementing an innovative chemical scaffold built on the 3- and 4-benzyloxy-benzylamino chemotype. Notably, compounds 28 (hBChE IC50 = 0.171 ± 0.063 μM) and 33 (hBChE IC50 = 0.167 ± 0.018 μM) emerged as top-ranked hBChE inhibitors. In silico simulations elucidated the binding modes of these compounds within hBChE. CNS availability was predicted using the BBB score algorithm, corroborated by in vitro permeability assessments with the most potent derivatives. Compound 33 was also inspected for aqueous solubility, microsomal and plasma stability. Chemoinformatics analysis validated these hBChE inhibitors for oral administration, indicating favorable gastrointestinal absorption in compliance with Lipinski's and Veber's rules. Safety assessments, crucial for the chronic administration typical in AD treatment, were conducted through cytotoxicity testing on human neuroblastoma (SH-SY5Y) and hepatocellular carcinoma (HepG2) cell lines.
Collapse
Affiliation(s)
- Filip Pidany
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacognosy and Pharmaceutical Botany, Charles University Akademika Heyrovskeho 1203 500 05 Hradec Kralove Czech Republic
| | - Jana Kroustkova
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacognosy and Pharmaceutical Botany, Charles University Akademika Heyrovskeho 1203 500 05 Hradec Kralove Czech Republic
| | - Jaroslav Jenco
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacognosy and Pharmaceutical Botany, Charles University Akademika Heyrovskeho 1203 500 05 Hradec Kralove Czech Republic
| | - Katerina Hradiska Breiterova
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacognosy and Pharmaceutical Botany, Charles University Akademika Heyrovskeho 1203 500 05 Hradec Kralove Czech Republic
| | - Lubica Muckova
- Biomedical Research Center, University Hospital Hradec Kralove Sokolska 581 500 05 Hradec Kralove Czech Republic
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Lucie Novakova
- Faculty of Pharmacy in Hradec Kralove, Department of Analytical Chemistry, Charles University Akademika Heyrovskeho 1203 500 05 Hradec Kralove Czech Republic
| | - Jiri Kunes
- Faculty of Pharmacy in Hradec Kralove, Department of Bioorganic and Organic Chemistry, Charles University Akademika Heyrovskeho 1203 500 05 Hradec Kralove Czech Republic
| | - Jakub Fibigar
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Tomas Kucera
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Martin Novak
- Biomedical Research Center, University Hospital Hradec Kralove Sokolska 581 500 05 Hradec Kralove Czech Republic
| | - Ales Sorf
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Martina Hrabinova
- Biomedical Research Center, University Hospital Hradec Kralove Sokolska 581 500 05 Hradec Kralove Czech Republic
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Lenka Pulkrabkova
- Biomedical Research Center, University Hospital Hradec Kralove Sokolska 581 500 05 Hradec Kralove Czech Republic
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Jiri Janousek
- Biomedical Research Center, University Hospital Hradec Kralove Sokolska 581 500 05 Hradec Kralove Czech Republic
| | - Ondrej Soukup
- Biomedical Research Center, University Hospital Hradec Kralove Sokolska 581 500 05 Hradec Kralove Czech Republic
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Daniel Jun
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove Sokolska 581 500 05 Hradec Kralove Czech Republic
- Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, University of Defence Trebesska 1575 500 01 Hradec Kralove Czech Republic
| | - Lucie Cahlikova
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacognosy and Pharmaceutical Botany, Charles University Akademika Heyrovskeho 1203 500 05 Hradec Kralove Czech Republic
| |
Collapse
|
12
|
Bartra C, Yuan Y, Vuraić K, Valdés-Quiroz H, Garcia-Baucells P, Slevin M, Pastorello Y, Suñol C, Sanfeliu C. Resveratrol Activates Antioxidant Protective Mechanisms in Cellular Models of Alzheimer's Disease Inflammation. Antioxidants (Basel) 2024; 13:177. [PMID: 38397775 PMCID: PMC10886200 DOI: 10.3390/antiox13020177] [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: 12/14/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Resveratrol is a natural phenolic compound with known benefits against neurodegeneration. We analyzed in vitro the protective mechanisms of resveratrol against the proinflammatory monomeric C-reactive protein (mCRP). mCRP increases the risk of AD after stroke and we previously demonstrated that intracerebral mCRP induces AD-like dementia in mice. Here, we used BV2 microglia treated with mCRP for 24 h in the presence or absence of resveratrol. Cells and conditioned media were collected for analysis. Lipopolysaccharide (LPS) has also been implicated in AD progression and so LPS was used as a resveratrol-sensitive reference agent. mCRP at the concentration of 50 µg/mL activated the nitric oxide pathway and the NLRP3 inflammasome pathway. Furthermore, mCRP induced cyclooxygenase-2 and the release of proinflammatory cytokines. Resveratrol effectively inhibited these changes and increased the expression of the antioxidant enzyme genes Cat and Sod2. As central mechanisms of defense, resveratrol activated the hub genes Sirt1 and Nfe2l2 and inhibited the nuclear translocation of the signal transducer NF-ĸB. Proinflammatory changes induced by mCRP in primary mixed glial cultures were also protected by resveratrol. This work provides a mechanistic insight into the protective benefits of resveratrol in preventing the risk of AD induced by proinflammatory agents.
Collapse
Affiliation(s)
- Clara Bartra
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (DIBAPS), 08036 Barcelona, Spain
- PhD Program in Biotechnology, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08034 Barcelona, Spain
| | - Yi Yuan
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Kristijan Vuraić
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Haydeé Valdés-Quiroz
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Pau Garcia-Baucells
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
| | - Mark Slevin
- School of Life Sciences, John Dalton Building, Manchester Metropolitan University, Manchester M15 6BH, UK;
- Centru Avansat de Cercetari Medicale si Farmaceutice (CCAMF), Universitatea de Medicina, Farmacie, Stiinte si Tehnologie “George Emil Palade” din Targu Mures, 540142 Targu Mures, Romania
| | - Ylenia Pastorello
- Department of Anatomy and Embryology, Universitatea de Medicina, Farmacie, Stiinte si Tehnologie “George Emil Palade” din Targu Mures, 540142 Targu Mures, Romania;
| | - Cristina Suñol
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (DIBAPS), 08036 Barcelona, Spain
| | - Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC, 08036 Barcelona, Spain; (C.B.); (Y.Y.); (K.V.); (H.V.-Q.); (P.G.-B.); (C.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (DIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
13
|
Tripathi MK, Ojha SK, Kartawy M, Khaliulin I, Hamoudi W, Amal H. Mutations associated with autism lead to similar synaptic and behavioral alterations in both sexes of male and female mouse brain. Sci Rep 2024; 14:10. [PMID: 38177238 PMCID: PMC10766975 DOI: 10.1038/s41598-023-50248-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/17/2023] [Indexed: 01/06/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder based on synaptic abnormalities. The estimated prevalence rate of male individuals diagnosed with ASD prevails over females is in a proportion of 4:1. Consequently, males remain the main focus in ASD studies in clinical and experimental settings. Meanwhile, some studies point to an underestimation of this disorder in females. In this work, we studied the sex differences of the synaptic and behavioral phenotypes of ASD mouse models. Juvenile male and female Shank3Δ4-22 and Cntnap2-/- mutant mice and their WT littermates were used in the experiments. The animals were subjected to a Three-Chamber Sociability Test, then euthanized, and the whole cortex was used for the evaluation of the synaptic phenotype. Protein levels of glutamatergic (NR1) and GABAergic (GAD1 and VGAT) neuronal markers were measured. Protein level of synaptophysin (Syp) was also measured. Dendritic spine density in somatosensory neurons was analyzed by Golgi staining methods. Spine Density and GAD1, NR1, VGAT, and Syp levels were significantly reduced in Shank3Δ4-22 and Cntnap2-/- mice compared to the control group irrespective of sex, indicating impaired synaptic development in the mutant mice. These results were consistent with the lack of differences in the three-chamber sociability test between male and female mice. In conclusion, female ASD mice of both mutations undergo similar synaptic aberrations as their male counterparts and need to be studied along with the male animals. Finally, this work urges the psychiatry scientific community to use both sexes in their investigations.
Collapse
Affiliation(s)
- Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
14
|
Kruglyakov D, Ojha SK, Kartawy M, Tripathi MK, Hamoudi W, Bazbaz W, Khaliulin I, Amal H. Nitric Oxide Synthase Inhibition Prevents Cell Proliferation in Glioblastoma. J Mol Neurosci 2023; 73:875-883. [PMID: 37843719 DOI: 10.1007/s12031-023-02166-3] [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: 09/07/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023]
Abstract
Glioblastoma multiforme (GBM) is a prevalent and aggressive primary brain tumor, presenting substantial treatment challenges and high relapse rates. GBM is characterized by alterations in molecular signaling and enzyme expression within malignant cells. This tumor exhibits elevated nitric oxide (NO.) levels. NO. is a crucial signaling molecule involved in the regulation of neuronal functions, synaptic transmission, and cell proliferation. It is primarily synthesized from L-arginine by nitric oxide synthase (NOS) enzymes. The increased levels of NO. in GBM stem from dysregulated activity and expression of clinically relevant NOS isoforms, particularly inducible NOS (iNOS) and neuronal NOS (nNOS). Based on this knowledge, we hypothesize that targeted pharmacological intervention with N6-(1-iminoethyl)-L-lysine (L-NIL), an iNOS inhibitor, and 7-Nitroindazole (7-NI), an nNOS inhibitor, may suggest a promising therapeutic strategy for the treatment of GBM. To test our hypothesis, we utilized the U87-MG cell line as an in vitro model of GBM. Our results showed that treatment with L-NIL and 7-NI led to a reduction in NO. levels, NOS activity, and clonogenic proliferation in U87-MG cells. These findings suggest that NO. and NOS enzymes might be prospective therapeutic targets for GBM.
Collapse
Affiliation(s)
- Daniel Kruglyakov
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wisam Bazbaz
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
15
|
Peng Q, Zeng Q, Wang F, Wu X, Zhang R, Shi G, Zhang M. Multi-engineered Graphene Extended-Gate Field-Effect Transistor for Peroxynitrite Sensing in Alzheimer's Disease. ACS NANO 2023; 17:21984-21992. [PMID: 37874899 DOI: 10.1021/acsnano.3c08499] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The expression of β-amyloid peptides (Aβ), a pathological indicator of Alzheimer's disease (AD), was reported to be inapparent in the early stage of AD. While peroxynitrite (ONOO-) is produced excessively and emerges earlier than Aβ plaques in the progression of AD, it is thus significant to sensitively detect ONOO- for early diagnosis of AD and its pathological research. Herein, we unveiled an integrated sensor for monitoring ONOO-, which consisted of a commercially available field-effect transistor (FET) and a high-performance multi-engineered graphene extended-gate (EG) electrode. In the configuration of the presented EG electrode, laser-induced graphene (LIG) intercalated with MnO2 nanoparticles (MnO2/LIG) can improve the electrical properties of LIG and the sensitivity of the sensor, and graphene oxide (GO)-MnO2/Hemin nanozyme with ONOO- isomerase activity can selectively trigger the isomerization of ONOO- to NO3-. With this synergistic effect, our EG-FET sensor can respond to the ONOO- with high sensitivity and selectivity. Moreover, taking advantage of our EG-FET sensor, we modularly assembled a portable sensing platform for wireless tracking ONOO- levels in the brain tissue of AD transgenic mice at earlier stages before massive Aβ plaques appeared, and we systematically explored the complex role of ONOO- in the occurrence and development of AD.
Collapse
Affiliation(s)
- Qiwen Peng
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Shanghai 200241, China
| | - Qiankun Zeng
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Shanghai 200241, China
| | - Fangbing Wang
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Shanghai 200241, China
| | - Xiaoyuan Wu
- School of Communication and Electric Engineering, East China Normal University, Shanghai 200241, China
| | - Runxi Zhang
- School of Communication and Electric Engineering, East China Normal University, Shanghai 200241, China
| | - Guoyue Shi
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Shanghai 200241, China
| | - Min Zhang
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Shanghai 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing 401120, China
| |
Collapse
|
16
|
Abdel-Haq M, Ojha SK, Hamoudi W, Kumar A, Tripathi MK, Khaliulin I, Domb AJ, Amal H. Effects of extended-release 7-nitroindazole gel formulation treatment on the behavior of Shank3 mouse model of autism. Nitric Oxide 2023; 140-141:41-49. [PMID: 37714296 DOI: 10.1016/j.niox.2023.09.003] [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: 08/20/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by behavioral deficits such as abnormalities in communication, social interaction, anxiety, and repetitive behavior. We have recently shown that the Shank3 mutation in mice representing a model of ASD causes excessive nitric oxide (NO) levels and aberrant protein S-nitrosylation. Further, 10-day daily injections of 7-NI, a neuronal nitric oxide synthase inhibitor, into Shank3Δ4-22 and Cntnap2(-/-) mutant mice (models of ASD) at a dose of 80 mg/kg reversed the manifestations of ASD phenotype. In this study, we proposed an extended release of 7-NI using a novel drug system. Importantly, unlike the intraperitoneal injections, our new preparation of poly (sebacic acid-co-ricinoleic acid) (PSARA) gel containing 7-NI was injected subcutaneously into the mutant mice only once. The animals underwent behavioral testing starting from day 3 post-injection. It should be noted that the developed PSARA gel formulation allowed a slow release of 7-NI maintaining the plasma level of the drug at ∼45 μg/ml/day. Further, we observed improved memory and social interaction and reduced anxiety-like behavior in Shank3 mutant mice. This was accompanied by a reduction in 3-nitrotyrosine levels (an indicator of nitrative/nitrosative stress) in plasma. Overall, we suggest that our single-dose formulation of PSARA gel is very efficient in rendering a therapeutic effect of 7-NI for at least 10 days. This approach may provide in the future a rational design of an effective ASD treatment using 7-NI and its clinical translation.
Collapse
Affiliation(s)
- Muhammad Abdel-Haq
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Awanish Kumar
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Abraham J Domb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel.
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel.
| |
Collapse
|
17
|
Liu L, Liu M, Zhang D, Song Z, Zhang H. DHT inhibits REDOX damage and neuroinflammation to reduce PND occurrence in aged mice via mmu_circ_0001442/miR-125a-3p/NUFIP2 axis. Brain Behav 2023; 13:e3180. [PMID: 37550899 PMCID: PMC10570480 DOI: 10.1002/brb3.3180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Perioperative neurocognitive disorder (PND) is the main cause of poor postoperative recovery in elderly patients with age-related reductions in androgen levels. However, the underlying mechanisms have not been completely elucidated. METHODS A mouse model of PND was constructed using abdominal surgery. Dihydrotestosterone (DHT), as the primary androgen, can improve the cognitive function of mice with PNDs by reducing REDOX damage. To clarify the role of circular RNA (circRNA) in DHT in improving cognitive function in mice with PND, circRNA sequencing was performed to analyze the expression of circRNA in the hippocampus of mice. RESULTS We confirmed that mmu_circ_0001442 is the primary circRNA responsive to DHT stimulation in mice with PND. The mmu_circ_0001442/miR-125a-3p/NUFIP2 axis was predicted and constructed according to the analysis of databases, including pita, miRanda, TargetScan, miRDB, micro-CDS, PolymiRTS, and TarBase v.8. Subsequently, the axis was verified by qPCR and double-luciferase reporter gene assays. In vitro, we found that DHT rarely had an effect on the growth of BV2 cells using the CCK-8 assay, but it attenuated the cytotoxic effect of lipopolysaccharide (LPS) on BV2 cells. In addition, we found that LPS stimulation promoted the release of proinflammatory cytokines, including IL-6 and TNF-α, in BV2 cells, whereas mmu_circ_0001442 knockdown and NUFIP2 knockdown partially abrogated this effect. CONCLUSIONS Taken together, DHT inhibited REDOX damage and neuroinflammation in the hippocampus to alleviate cognitive disorders in mice with PNDs via activation of the mmu_circ_0001442/miR-125a-3p/NUFIP2 axis. This study provides a novel rationale for developing DHT as a potential therapeutic agent for PND prevention.
Collapse
Affiliation(s)
- Li Liu
- Department of OncologyJiangxi Provincial People's HospitalNanchangJiangxiP. R. China
| | - Mei Liu
- Department of AnesthesiologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Daying Zhang
- Department of Pain ManagementThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Zhiping Song
- Department of AnesthesiologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Huaigen Zhang
- Department of AnesthesiologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| |
Collapse
|
18
|
Inácio Â, Aguiar L, Rodrigues B, Pires P, Ferreira J, Matos A, Mendonça I, Rosa R, Bicho M, Medeiros R, Bicho MC. Genetic Modulation of HPV Infection and Cervical Lesions: Role of Oxidative Stress-Related Genes. Antioxidants (Basel) 2023; 12:1806. [PMID: 37891885 PMCID: PMC10604255 DOI: 10.3390/antiox12101806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Human papillomavirus (HPV) infection is a necessary but not sufficient factor for the development of invasive cervical cancer (ICC) and high-grade intraepithelial lesion (HSIL). Oxidative stress is known to play a crucial role in HPV infection and carcinogenesis. In this study, we comprehensively investigate the modulation of HPV infection, HSIL and ICC, and ICC through an exploration of oxidative stress-related genes: CβS, MTHFR, NOS3, ACE1, CYBA, HAP, ACP1, GSTT1, GSTM1, and CYP1A1. Notably, the ACE1 gene emerges as a prominent factor with the presence of the I allele offering protection against HPV infection. The association of NOS3 with HPV infection is perceived with the 4a allele showing a protective effect. The presence of the GSTT1 null mutant correlates with increased susceptibility to HPV infection, HSIL and ICC, and ICC. This study also uncovers intriguing epistatic interactions among some of the genes that further accentuate their roles in disease modulation. Indeed, the epistatic interactions between the BB genotype (ACP1) and DD genotype (ECA1) were shown to increase the risk of HPV infection, and the interaction between BB (ACP1) and 0.0 (GSTT1) was associated with HPV infection and cervical lesions. These findings underscore the pivotal role of four oxidative stress-related genes in HPV-associated cervical lesions and cancer development, enriching our clinical understanding of the genetic influences on disease manifestation. The awareness of these genetic variations holds potential clinical implications.
Collapse
Affiliation(s)
- Ângela Inácio
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto de Saúde Ambiental (ISAMB) e Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto Bento da Rocha Cabral, 1250-047 Lisboa, Portugal
| | - Laura Aguiar
- Instituto de Saúde Ambiental (ISAMB) e Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto Bento da Rocha Cabral, 1250-047 Lisboa, Portugal
| | - Beatriz Rodrigues
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Patrícia Pires
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Joana Ferreira
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto de Saúde Ambiental (ISAMB) e Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto Bento da Rocha Cabral, 1250-047 Lisboa, Portugal
| | - Andreia Matos
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto de Saúde Ambiental (ISAMB) e Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto Bento da Rocha Cabral, 1250-047 Lisboa, Portugal
| | - Inês Mendonça
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Raquel Rosa
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Manuel Bicho
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto de Saúde Ambiental (ISAMB) e Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto Bento da Rocha Cabral, 1250-047 Lisboa, Portugal
| | - Rui Medeiros
- Molecular Oncology & Viral Pathology Group, Research Center (CI-IPOP)/RISE@CI-IPOP, Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal
| | - Maria Clara Bicho
- Instituto de Saúde Ambiental (ISAMB) e Laboratório Associado TERRA, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto de Medicina Preventiva e Saúde Pública, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
19
|
Jian J, Li LG, Zhao PJ, Zheng RJ, Dong XW, Zhao YH, Yin BQ, Li S, Cheng H, Li HL, Li EY. Mouse nerve growth factor suppresses neuronal apoptosis in valproic acid-induced autism spectrum disorder rats by regulating the phosphoinositide-3-kinase/serine/threonine kinase signaling pathway. Pharmacogenet Genomics 2023; 33:101-110. [PMID: 37261937 DOI: 10.1097/fpc.0000000000000498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by deficits in social communication and restrictive behaviors. Mouse nerve growth factor (mNGF), a neurotrophic factor, is critical for neuronal growth and survival, and the mNGF treatment is considered a promising therapy for neurodegeneration. In light of this, we aimed to evaluate the effect of mNGF on neurological function in ASD. METHODS An ASD rat model was established by intraperitoneal injection of valproic acid (VPA). Social behavior, learning, and memory of the rats were measured. TdT-mediated dUTP Nick-end labeling and Nissl assays were performed to detect neuronal apoptosis and survival in the hippocampus and prefrontal cortex. Apoptosis-related proteins and oxidative stress markers were detected. RESULTS mNGF improved locomotor activity, exploratory behavior, social interaction, and spatial learning and memory in VPA-induced ASD rats. In the hippocampus and prefrontal cortex, mNGF suppressed neuronal apoptosis, increased the number of neurons, superoxide dismutase, and glutathione levels, and decreased reactive oxygen species, nitric oxide, TNF-α, and IL-1β levels compared with the VPA group. In addition, mNGF increased the levels of Bcl-2, p-phosphoinositide-3-kinase (PI3K), and p-serine/threonine kinase (Akt), and decreased the levels of Bax and cleaved caspase-3, while the PI3K inhibitor LY294002 reversed these effects. CONCLUSION These data suggest that mNGF suppressed neuronal apoptosis and ameliorated the abnormal behaviors in VPA-induced ASD rats, in part, by activating the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Jie Jian
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - Li-Guo Li
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
- Institute of Health Engineering, Zhengzhou Health Vocational College, Zhengzhou
| | - Peng-Ju Zhao
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - Rui-Juan Zheng
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - Xian-Wen Dong
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - Yong-Hong Zhao
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - Bao-Qi Yin
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - Sheng Li
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Hui Cheng
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - Hong-Lei Li
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| | - En-Yao Li
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, the Fifth Affiliated Hospital of Zhengzhou University
| |
Collapse
|
20
|
Zhang L, Chen L, Qi M, Yu F, Ni X, Hong H, Xu H, Xu S. Glyphosate induces autophagy in hepatic L8824 cell line through NO-mediated activation of RAS/RAF/MEK/ERK signaling pathway and energy metabolism disorders. FISH & SHELLFISH IMMUNOLOGY 2023; 137:108772. [PMID: 37100311 DOI: 10.1016/j.fsi.2023.108772] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/22/2023]
Abstract
Glyphosate is an herbicide commonly used worldwide, and its substantial use causes widespread pollution with runoff. However, research on glyphosate toxicity has mostly remained at the embryonic level and existing studies are limited. In the present study, we investigated whether glyphosate can induce autophagy in hepatic L8824 cells by regulating energy metabolism and rat sarcoma (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated extracellular signal-regulated kinase (MEK)/extracellular regulated protein kinases (ERK) signaling by activating nitric oxide (NO). First, we selected 0, 50, 200, and 500 μg/mL as the challenge doses, according to the inhibitory concentration of 50% (IC50) of glyphosate. The results showed that glyphosate exposure increased the enzyme activity of inducible nitric oxide synthase (iNOS), which in turn increased the NO content. The activity and expression of enzymes related to energy metabolism, such as hexokinase (HK)1, HK2, phosphofructokinase (PFK), phosphokinase (PK), succinate dehydrogenase (SDH), and nicotinamide adenine dinucleotide with hydrogen (NADH), were inhibited, and the RAS/RAF/MEK/ERK signaling pathway was activated. This led to the negative expression of mammalian target of rapamycin (mTOR) and P62 in hepatic L8824 cells and the activation of the autophagy marker genes microtubule-associated proteins light chain 3 (LC3) and Beclin1 to induce autophagy. The above results were dependent on glyphosate concentration. To verify whether autophagy can be excited by the RAS/RAF/MEK/ERK signaling pathway, we treated L8824 cells with the ERK inhibitor U0126 and found that the autophagy gene LC3 was reduced due to the inhibition of ERK, thus demonstrating the reliability of the results. In conclusion, our results demonstrate that glyphosate can induce autophagy in hepatic L8824 cells by activating NO, thus regulating energy metabolism and the RAS/RAF/MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Linlin Zhang
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Lu Chen
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Meng Qi
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Fuchang Yu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Xiaotong Ni
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Haozheng Hong
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Haotian Xu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China.
| | - Shiwen Xu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China; Key Laboratory of Tarim Animal Husbandry Technology Corps, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China.
| |
Collapse
|
21
|
Amal H, Steinert JR. Editorial: Nitric oxide signaling from synapse to disease. Free Radic Biol Med 2023; 196:9-10. [PMID: 36626954 DOI: 10.1016/j.freeradbiomed.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham, NG7 2NR, UK.
| |
Collapse
|