1
|
Verlinden SF. The genetic advantage of healthy centenarians: unraveling the central role of NLRP3 in exceptional healthspan. FRONTIERS IN AGING 2024; 5:1452453. [PMID: 39301197 PMCID: PMC11410711 DOI: 10.3389/fragi.2024.1452453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Despite extensive research into extending human healthspan (HS) and compressing morbidity, the mechanisms underlying aging remain elusive. However, a better understanding of the genetic advantages responsible for the exceptional HS of healthy centenarians (HC), who live in good physical and mental health for one hundred or more years, could lead to innovative health-extending strategies. This review explores the role of NLRP3, a critical component of innate immunity that significantly impacts aging. It is activated by pathogen-associated signals and self-derived signals that increase with age, leading to low-grade inflammation implicated in age-related diseases. Furthermore, NLRP3 functions upstream in several molecular aging pathways, regulates cellular senescence, and may underlie the robust health observed in HC. By targeting NLRP3, mice exhibit a phenotype akin to that of HC, the HS of monkeys is extended, and aging symptoms are reversed in humans. Thus, targeting NLRP3 could offer a promising approach to extend HS. Additionally, a paradigm shift is proposed. Given that the HS of the broader population is 30 years shorter than that of HC, it is postulated that they suffer from a form of accelerated aging. The term 'auto-aging' is suggested to describe accelerated aging driven by NLRP3.
Collapse
|
2
|
Shi Z, Li H, Cheng J, Zhang W, Ruan J, Zhang Q, Dang Z, Zhang Y, Wang T. Constituents from Dolichos lablab L. Flowers and Their Anti-Inflammatory Effects via Inhibition of IL-1β Release. Molecules 2024; 29:3751. [PMID: 39202831 PMCID: PMC11357617 DOI: 10.3390/molecules29163751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
The occurrence of inflammation is closely related to the activation of the NLRP3 inflammasome. IL-1β produced during the activation of the NLRP3 inflammasome has strong pro-inflammatory activity and can also promote the release of inflammatory factors by other immune cells, exacerbating inflammatory damage to tissues. Utilizing IL-1β as the detection index to find small-molecule inhibitors targeting NLRP3 from natural products will benefit the search for drugs for inflammation-related diseases. During the exploration of anti-inflammatory active components derived from the flowers of Dolichos lablab L., an ingredient in traditional Chinese medicine with dual applications in both medicinal treatment and dietary consumption, fourteen compounds (1-14), including seven previously unreported ones, named flosdolilabnitrogenousols A-D (1-4) and flosdolilabsaponins A-C (5-7), were found. Their structures were established through extensive NMR spectra determination, HR-ESI-MS analysis, ECD calculations, and chemical reactions. Flosdolilabsaponin A (5) stands out as an exceptionally rare tetracyclic lactone oleane-type saponin. Additionally, the inhibitory activity on IL-1β release of all compounds, without cytotoxicity, was evaluated using BMDMs stimulated with LPS/Nigericin. An Elisa assay revealed that compounds 1, 8, 9, and 11-14 exhibited significant inhibition of IL-1β release at a concentration of 30 μM. Structure-activity relationships were also discussed. This study indicates that D. lablab flowers possess anti-inflammatory activity, which might exert its effect by suppressing the activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Zhongwei Shi
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (Z.S.); (H.L.)
| | - Huimin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (Z.S.); (H.L.)
| | - Jiaming Cheng
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (J.C.); (W.Z.); (J.R.); (Q.Z.); (Z.D.)
| | - Wei Zhang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (J.C.); (W.Z.); (J.R.); (Q.Z.); (Z.D.)
| | - Jingya Ruan
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (J.C.); (W.Z.); (J.R.); (Q.Z.); (Z.D.)
| | - Qianqian Zhang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (J.C.); (W.Z.); (J.R.); (Q.Z.); (Z.D.)
| | - Zhunan Dang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (J.C.); (W.Z.); (J.R.); (Q.Z.); (Z.D.)
| | - Yi Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (Z.S.); (H.L.)
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (J.C.); (W.Z.); (J.R.); (Q.Z.); (Z.D.)
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (Z.S.); (H.L.)
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China; (J.C.); (W.Z.); (J.R.); (Q.Z.); (Z.D.)
| |
Collapse
|
3
|
Bonam SR, Mastrippolito D, Georgel P, Muller S. Pharmacological targets at the lysosomal autophagy-NLRP3 inflammasome crossroads. Trends Pharmacol Sci 2024; 45:81-101. [PMID: 38102020 DOI: 10.1016/j.tips.2023.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023]
Abstract
Many aspects of cell homeostasis and integrity are maintained by the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome. The NLRP3 oligomeric protein complex assembles in response to exogenous and endogenous danger signals. This inflammasome has also been implicated in the pathogenesis of a range of disease conditions, particularly chronic inflammatory diseases. Given that NLRP3 modulates autophagy, which is also a key regulator of inflammasome activity, excessive inflammation may be controlled by targeting this intersecting pathway. However, specific niche areas of NLRP3-autophagy interactions and their reciprocal regulatory mechanisms remain underexplored. Consequently, we lack treatment methods specifically targeting this pivotal axis. Here, we discuss the potential of such strategies in the context of autoimmune and metabolic diseases and propose some research avenues.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Dylan Mastrippolito
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France
| | - Philippe Georgel
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; Strasbourg Institute of Drug Discovery and Development (IMS), Strasbourg, France; Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France; University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.
| |
Collapse
|
4
|
Kaffe D, Kaplanis SI, Karagogeos D. The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination. Curr Issues Mol Biol 2023; 45:9526-9548. [PMID: 38132442 PMCID: PMC10742427 DOI: 10.3390/cimb45120596] [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: 10/02/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The dysfunction of myelinating glial cells, the oligodendrocytes, within the central nervous system (CNS) can result in the disruption of myelin, the lipid-rich multi-layered membrane structure that surrounds most vertebrate axons. This leads to axonal degeneration and motor/cognitive impairments. In response to demyelination in the CNS, the formation of new myelin sheaths occurs through the homeostatic process of remyelination, facilitated by the differentiation of newly formed oligodendrocytes. Apart from oligodendrocytes, the two other main glial cell types of the CNS, microglia and astrocytes, play a pivotal role in remyelination. Following a demyelination insult, microglia can phagocytose myelin debris, thus permitting remyelination, while the developing neuroinflammation in the demyelinated region triggers the activation of astrocytes. Modulating the profile of glial cells can enhance the likelihood of successful remyelination. In this context, recent studies have implicated autophagy as a pivotal pathway in glial cells, playing a significant role in both their maturation and the maintenance of myelin. In this Review, we examine the role of substances capable of modulating the autophagic machinery within the myelinating glial cells of the CNS. Such substances, called caloric restriction mimetics, have been shown to decelerate the aging process by mitigating age-related ailments, with their mechanisms of action intricately linked to the induction of autophagic processes.
Collapse
Affiliation(s)
- Despoina Kaffe
- Department of Biology, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
| | - Stefanos Ioannis Kaplanis
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| | - Domna Karagogeos
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| |
Collapse
|
5
|
Li Q, Zhang Q, Kim YR, Gaddam RR, Jacobs JS, Bachschmid MM, Younis T, Zhu Z, Zingman L, London B, Rauckhorst AJ, Taylor EB, Norris AW, Vikram A, Irani K. Deficiency of endothelial sirtuin1 in mice stimulates skeletal muscle insulin sensitivity by modifying the secretome. Nat Commun 2023; 14:5595. [PMID: 37696839 PMCID: PMC10495425 DOI: 10.1038/s41467-023-41351-1] [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: 11/12/2021] [Accepted: 08/31/2023] [Indexed: 09/13/2023] Open
Abstract
Downregulation of endothelial Sirtuin1 (Sirt1) in insulin resistant states contributes to vascular dysfunction. Furthermore, Sirt1 deficiency in skeletal myocytes promotes insulin resistance. Here, we show that deletion of endothelial Sirt1, while impairing endothelial function, paradoxically improves skeletal muscle insulin sensitivity. Compared to wild-type mice, male mice lacking endothelial Sirt1 (E-Sirt1-KO) preferentially utilize glucose over fat, and have higher insulin sensitivity, glucose uptake, and Akt signaling in fast-twitch skeletal muscle. Enhanced insulin sensitivity of E-Sirt1-KO mice is transferrable to wild-type mice via the systemic circulation. Endothelial Sirt1 deficiency, by inhibiting autophagy and activating nuclear factor-kappa B signaling, augments expression and secretion of thymosin beta-4 (Tβ4) that promotes insulin signaling in skeletal myotubes. Thus, unlike in skeletal myocytes, Sirt1 deficiency in the endothelium promotes glucose homeostasis by stimulating skeletal muscle insulin sensitivity through a blood-borne mechanism, and augmented secretion of Tβ4 by Sirt1-deficient endothelial cells boosts insulin signaling in skeletal muscle cells.
Collapse
Affiliation(s)
- Qiuxia Li
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA.
| | - Quanjiang Zhang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Young-Rae Kim
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Ravinder Reddy Gaddam
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Julia S Jacobs
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | | | - Tsneem Younis
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Zhiyong Zhu
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Veterans Affairs Medical Center, Iowa City, IA, 52242, USA
| | - Leonid Zingman
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Veterans Affairs Medical Center, Iowa City, IA, 52242, USA
| | - Barry London
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Adam J Rauckhorst
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- FOEDRC Metabolomics Core Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Eric B Taylor
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- FOEDRC Metabolomics Core Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Andrew W Norris
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- FOEDRC Metabolic Phenotyping Core Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Ajit Vikram
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kaikobad Irani
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Veterans Affairs Medical Center, Iowa City, IA, 52242, USA.
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
6
|
Sidor K, Jeznach A, Hoser G, Skirecki T. 1-Methylnicotinamide (1-MNA) inhibits the activation of the NLRP3 inflammasome in human macrophages. Int Immunopharmacol 2023; 121:110445. [PMID: 37290319 DOI: 10.1016/j.intimp.2023.110445] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
The NLRP3 inflammasome is among the most potent intracellular sensors of danger and disturbances of cellular homeostasis that can lead to the release of IL-1β and cell death, or pyroptosis. Despite its protective role, this mechanism is involved in the pathogenesis of numerous inflammatory diseases; therefore, it is seen as a potential therapeutic target. 1-methylnicotinamide (1-MNA) is a direct metabolite of nicotinamide and was previously shown to display several immunomodulatory properties, including a reduction in the reactive oxygen species (ROS). Here, we investigated whether 1-MNA could influence the activation of the NLRP3 inflammasome in human macrophages. In differentiated human macrophages we observed that 1-MNA specifically reduced the activation of the NLRP3 inflammasome. This effect was related to the scavenging of ROS, as exogenous H2O2 was able to restore NLRP3 activation. Additionally, 1-MNA increased the mitochondrial membrane potential, indicating that it did not inhibit oxidative phosphorylation. Moreover, at high but not low concentrations, 1-MNA decreased NF-κB activation and the level of pro-IL-1β. Interestingly, 1-MNA did not reduce the secretion of IL-6 upon endotoxin stimulation, confirming that its primary immunomodulatory effect on human macrophages is dependent on the NLRP3 inflammasome. Taken together, we have shown for the first time that 1-MNA reduced the activation of the NLRP3 inflammasome in human macrophages via an ROS-dependent pathway. Our results indicate a novel potential use of 1-MNA in NLRP3-related disorders.
Collapse
Affiliation(s)
- Karolina Sidor
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Aldona Jeznach
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Grażyna Hoser
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
7
|
Hosseini E, Kohan-Ghadr HR, Bazrafkan M, Amorim CA, Askari M, Zakeri A, Mousavi SN, Kafaeinezhad R, Afradiasbagharani P, Esfandyari S, Nazari M. Rescuing fertility during COVID-19 infection: exploring potential pharmacological and natural therapeutic approaches for comorbidity, by focusing on NLRP3 inflammasome mechanism. J Assist Reprod Genet 2023; 40:1173-1185. [PMID: 36892705 PMCID: PMC9995769 DOI: 10.1007/s10815-023-02768-1] [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: 12/08/2022] [Accepted: 02/28/2023] [Indexed: 03/10/2023] Open
Abstract
The respiratory system was primarily considered the only organ affected by Coronavirus disease 2019 (COVID-19). As the pandemic continues, there is an increasing concern from the scientific community about the future effects of the virus on male and female reproductive organs, infertility, and, most significantly, its impact on the future generation. The general presumption is that if the primary clinical symptoms of COVID-19 are not controlled, we will face several challenges, including compromised infertility, infection-exposed cryopreserved germ cells or embryos, and health complications in future generations, likely connected to the COVID-19 infections of parents and ancestors. In this review article, we dedicatedly studied severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) virology, its receptors, and the effect of the virus to induce the activation of inflammasome as the main arm of the innate immune response. Among inflammasomes, nucleotide oligomerization domain-like receptor protein, pyrin domain containing 3 (NLRP3) inflammasome pathway activation is partly responsible for the inflicted damages in both COVID-19 infection and some reproductive disorders, so the main focus of the discussion is on NLRP3 inflammasome in the pathogenesis of COVID-19 infection alongside in the reproductive biology. In addition, the potential effects of the virus on male and female gonad functions were discussed, and we further explored the potential natural and pharmacological therapeutic approaches for comorbidity via NLRP3 inflammasome neutralization to develop a hypothesis for averting the long-term repercussions of COVID-19. Since activation of the NLRP3 inflammasome pathway contributes to the damage caused by COVID-19 infection and some reproductive disorders, NLRP3 inflammasome inhibitors have a great potential to be considered candidates for alleviating the pathological effects of the COVID-19 infection on the germ cells and reproductive tissues. This would impede the subsequent massive wave of infertility that may threaten the patients.
Collapse
Affiliation(s)
- Elham Hosseini
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Obstetrics and Gynecology, Mousavi Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamid-Reza Kohan-Ghadr
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI USA
| | - Mahshid Bazrafkan
- Reproductive Biotechnology Research Center, Avicenna Research Institute (ARI), ACECR, Tehran, Iran
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maryam Askari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Armin Zakeri
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyedeh Neda Mousavi
- Department of Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Raheleh Kafaeinezhad
- Department of Biology, Faculty of Basic Sciences, University of Maragheh, Maragheh, Iran
| | | | - Sahar Esfandyari
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Mahboobeh Nazari
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
8
|
He J, Deng Y, Ren L, Jin Z, Yang J, Yao F, Liu Y, Zheng Z, Chen D, Wang B, Zhang Y, Nan G, Wang W, Lin R. Isoliquiritigenin from licorice flavonoids attenuates NLRP3-mediated pyroptosis by SIRT6 in vascular endothelial cells. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115952. [PMID: 36442759 DOI: 10.1016/j.jep.2022.115952] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGIC RELEVANCE Licorice is a traditional Chinese medicine that has been used for cardiovascular diseases. Recent studies found that supplementation with licorice extracts attenuated the development of atherosclerosis (AS) in hypercholesterolemic patients. Many studies have shown that licorice flavonoids, the main active components of licorice, have a variety of pharmacological effects, including anti-inflammation, regulation of lipid metabolism, and antioxidation. However, the key active components against AS in licorice flavonoids are still unclear. AIM OF THE STUDY The aim of this paper is to investigate the active components of licorice flavonoids that exert anti-atherosclerotic effects and the underlying mechanisms. MATERIALS AND METHODS Network pharmacology was used to screen the active components of licorice flavonoids that have anti-atherosclerotic effects. Combining bioinformatics analysis and in vitro studies, the effects and underlying mechanisms of the active component isoliquiritigenin (ISL) on cell pyroptosis were further investigated in tumor necrosis factor (TNF)-α-treated human umbilical vein endothelial cells (HUVECs). RESULTS We constructed a compound-target network and screened 3 active components, namely, ISL, glabridin, and naringenin in licorice flavonoids. The half maximal effective concentration values of these 3 components suggested that ISL was the key active component against TNF-α-induced endothelial cell injury. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that ISL could potentially treat AS via the nucleotide-binding and oligomerization domain (NOD)-like receptor signaling pathway. An in vitro study verified that ISL suppressed TNF-α-induced NLRP3 activation and pyroptosis in HUVECs. The molecular docking and cellular thermal shift assay showed good compatibility between ISL and class III histone deacetylase sirtuin 6 (SIRT6). Moreover, we found that ISL upregulated the expression of SIRT6 in TNF-α-treated HUVECs. Further study found that SIRT6 knockdown reduced the inhibitory effect of ISL on pyroptosis, whereas the NLRP3 inhibitor reversed this process in TNF-α-treated HUVECs. CONCLUSIONS Our results demonstrate that ISL is a key active component of licorice flavonoids. ISL attenuates NLRP3-mediated vascular endothelial cell pyroptosis via SIRT6, and SIRT6 may be a potential target of ISL for the treatment of AS.
Collapse
Affiliation(s)
- Jianyu He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Ying Deng
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Lingxuan Ren
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Zhen Jin
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jianjun Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Feng Yao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yizhen Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Zihan Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Danli Chen
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Bo Wang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yirong Zhang
- Department of Medical Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Guanjun Nan
- School of Pharmacy, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Weirong Wang
- Department of Medical Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| | - Rong Lin
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
9
|
Wu Y, Pi D, Zhou S, Wang W, Ye H, Yi Z, Chen Y, Ouyang M. Yiqi Chutan Formula Reverses Cisplatin-Induced Apoptosis and Ferroptosis of Skeletal Muscle by Alleviating Oxidative Stress. Integr Cancer Ther 2023; 22:15347354231172117. [PMID: 37132527 PMCID: PMC10161340 DOI: 10.1177/15347354231172117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Cisplatin is a widely used anticancer drug in clinic, but it has a damaging effect on skeletal muscle cells. Clinical observation showed that Yiqi Chutan formula (YCF) had a alleviating effect on cisplatin toxicity. METHODS In vitro cell model and in vivo animal model were used to observe the damage effect of cisplatin on skeletal muscle cells and verify that YCF reversed cisplatin induced skeletal muscle damage. The levels of oxidative stress, apoptosis and ferroptosis were measured in each group. RESULTS Both in vitro and in vivo studies have confirmed that cisplatin increases the level of oxidative stress in skeletal muscle cells, thus inducing cell apoptosis and ferroptosis. YCF treatment can effectively reverse cisplatin induced oxidative stress in skeletal muscle cells, thereby alleviating cell apoptosis and ferroptosis, and ultimately protecting skeletal muscle. CONCLUSIONS YCF reversed cisplatin-induced apoptosis and ferroptosis of skeletal muscle by alleviating oxidative stress.
Collapse
Affiliation(s)
- Yingchao Wu
- Jinan University, Guangzhou, Guangdong, China
| | - Dajin Pi
- Jinan University, Guangzhou, Guangdong, China
| | - Shuyao Zhou
- Guangdong Hanchao Traditional Chinese Medicine Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Wuhong Wang
- Jinan University, Guangzhou, Guangdong, China
| | - Huan Ye
- Jinan University, Guangzhou, Guangdong, China
| | - Zhongjia Yi
- Jinan University, Guangzhou, Guangdong, China
| | - Yiliu Chen
- Jinan University, Guangzhou, Guangdong, China
| | | |
Collapse
|
10
|
Zhou J, Yang R, Sun Y, Luo F, Zhang J, Ma H, Guan M. HClO-triggered interventional probe enabled early detection and intervention of atherosclerosis. Analyst 2022; 148:163-174. [PMID: 36464987 DOI: 10.1039/d2an01374f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of early atherosclerosis (AS). Targeting foam cell formation can be a promising approach for the early detection and prevention of AS. However, only a few studies have actually examined foam cells in vivo, and most methods combined nanotechnology with angiography, which is complex and could cause further damage to the endothelium. Herein, based on methylene blue, a biosafe NIR dye approved by the FDA, an interventional probe (HMB-NA@Mp) triggered by hypochlorous acid (HClO) was designed for imaging foam cells easily, safely, and effectively in the early stage of AS. Here, encapsulation of the probe by foam cells targeted platelet membrane (Mp) increased probe targeting and reduced toxicity. Cell and animal experimental results showed that the probe could accumulate at the lesion site and significantly enhance fluorescence in the early AS model group. Remarkably, at the same time, it could also release the metabolite niacin, which played a role in inhibiting atherosclerosis. Thus, HMB-NA@Mp is expected to be a powerful means for the early detection and timely intervention of early AS in the absence of clinical symptoms.
Collapse
Affiliation(s)
- Jie Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Ruhe Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Yiwen Sun
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Fusui Luo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Jin Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Huili Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Min Guan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
11
|
Zhang M, Li Y, Guo Y, Xu J. Arginine Regulates NLRP3 Inflammasome Activation Through SIRT1 in Vascular Endothelial Cells. Inflammation 2021; 44:1370-1380. [PMID: 33630211 DOI: 10.1007/s10753-021-01422-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/23/2020] [Accepted: 01/18/2021] [Indexed: 12/11/2022]
Abstract
L-arginine (Arg), a semi-essential amino acid, has recently been shown to attenuate inflammatory response during cardiovascular disease. NLRP3 inflammasome serves a central role in amplification of cellular inflammation. In this study, we aimed to confirm the modulatory effect of Arg on NLRP3 inflammasome and the underlying mechanisms in vascular endothelial cells (ECs). Arg suppressed NLRP3 inflammasome activation in ECs stimulated with lipopolysaccharide (LPS) and adenosine triphosphate (ATP). Moreover, treatment with Arg increased the expression of the deacetylase sirtuin 1 (SIRT1) in ECs. Importantly, knockdown of SIRT1 abolished the inhibitory potential of Arg on the activation of NLRP3 inflammasome. Further study indicated that Arg also alleviated LPS plus ATP-induced the generation of reactive oxygen species (ROS) in ECs. In addition, Arg may regulate NLRP3 inflammasome activation partly through suppression of ROS production. In combination, we speculate that Arg exerts an inhibitory effect on the activation of NLRP3 inflammasome in ECs, which may be partly mediated by SIRT1 and ROS.
Collapse
Affiliation(s)
| | - Yanxiang Li
- School of Pharmacy, Weifang Medical University, Weifang, China
- School of Pharmacy, Taizhou Polytechnic College, Taizhou, China
| | - Yujie Guo
- School of Medicine, Nantong University, Nantong, China.
| | - Jiashuo Xu
- School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
12
|
Arenas-Jal M, Suñé-Negre JM, García-Montoya E. Therapeutic potential of nicotinamide adenine dinucleotide (NAD). Eur J Pharmacol 2020; 879:173158. [PMID: 32360833 DOI: 10.1016/j.ejphar.2020.173158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/06/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022]
Abstract
Nicotinamide adenine nucleotide (NAD) is a small ubiquitous hydrophilic cofactor that participates in several aspects of cellular metabolism. As a coenzyme it has an essential role in the regulation of energetic metabolism, but it is also a cosubstrate for enzymes that regulate fundamental biological processes such as transcriptional regulation, signaling and DNA repairing among others. The fluctuation and oxidative state of NAD levels regulate the activity of these enzymes, which is translated into marked effects on cellular function. While alterations in NAD homeostasis are a common feature of different conditions and age-associated diseases, in general, increased NAD levels have been associated with beneficial health effects. Due to its therapeutic potential, the interest in this molecule has been renewed, and the regulation of NAD metabolism has become an attractive target for drug discovery. In fact, different approaches to replenish or increase NAD levels have been tested, including enhancement of biosynthesis and inhibition of NAD breakdown. Despite further research is needed, this review provides an overview and update on NAD metabolism, including the therapeutic potential of its regulation, as well as pharmacokinetics, safety, precautions and formulation challenges of NAD supplementation.
Collapse
Affiliation(s)
- Marta Arenas-Jal
- Pharmacy and Pharmaceutical Technology Department (Faculty of Pharmacy and Food Sciences), University of Barcelona, Barcelona, Spain; ICN2 - Catalan Institute of Nanoscience and Nanotechnology (Autonomous University of Barcelona), Bellaterra (Barcelona), Spain.
| | - J M Suñé-Negre
- Pharmacy and Pharmaceutical Technology Department (Faculty of Pharmacy and Food Sciences), University of Barcelona, Barcelona, Spain
| | - Encarna García-Montoya
- Pharmacy and Pharmaceutical Technology Department (Faculty of Pharmacy and Food Sciences), University of Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Abstract
Niacin (nicotinic acid) is a potent lipid-lowering agent that has been used for prevention of coronary heart disease. Niacin activates the HCAR2 receptor found on adipocytes, macrophages and various immune cells throughout the body. Activation of the HCAR2 receptor by niacin results in beneficial anti-inflammatory effects that are independent of lipid lowering. This review summarizes the use of niacin in treatment of dyslipidemia, the pharmacogenetics of niacin response and the potential role of HCAR2 signaling in the treatment of a variety of inflammatory and metabolic diseases.
Collapse
Affiliation(s)
- Sony Tuteja
- Department of Medicine, Division of Translational Medicine & Human Genetics, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, 11-143, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
He Y, Yang G, Yao F, Xian Y, Wang G, Chen L, Lv X, Gao H, Zheng Z, Sun L, Wang W, Lin R. Sitagliptin inhibits vascular inflammation via the SIRT6-dependent signaling pathway. Int Immunopharmacol 2019; 75:105805. [DOI: 10.1016/j.intimp.2019.105805] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
|
15
|
Zhou X, Wu Y, Ye L, Wang Y, Zhang K, Wang L, Huang Y, Wang L, Xian S, Zhang Y, Chen Y. Aspirin alleviates endothelial gap junction dysfunction through inhibition of NLRP3 inflammasome activation in LPS-induced vascular injury. Acta Pharm Sin B 2019; 9:711-723. [PMID: 31384532 PMCID: PMC6664043 DOI: 10.1016/j.apsb.2019.02.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/04/2019] [Accepted: 01/11/2019] [Indexed: 12/22/2022] Open
Abstract
The loss of endothelial connective integrity and endothelial barrier dysfunction can lead to increased vascular injury, which is related to the activation of endothelial inflammasomes. There are evidences that low concentrations of aspirin can effectively prevent cardiovascular diseases. We hypothesized that low-dose aspirin could ameliorate endothelial injury by inhibiting the activation of NLRP3 inflammasomes and ultimately prevent cardiovascular diseases. Microvascular endothelial cells were stimulated by lipopolysaccharide (2 μg/mL) and administrated by 0.1–2 mmol/L aspirin. The wild type mice were stimulated with LPS (100 μg/kg/day), and 1 h later treated with aspirin (12.5, 62.5, or 125 mg/kg/day) and dexamethasone (0.0182 mg/kg/day) for 7 days. Plasma and heart were harvested for measurement of ELISA and immunofluorescence analyses. We found that aspirin could inhibit NLRP3 inflammasome formation and activation in vitro in dose-dependent manner and has correlation between the NLRP3 inflammasome and the ROS/TXNIP pathway. We also found that low-concentration aspirin could inhibit the formation and activation of NLRP3 inflammasome and restore the expression of the endothelial tight junction protein zonula occludens-1/2 (ZO1/2). We assume that aspirin can ameliorate the endothelial layer dysfunction by suppressing the activation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xing Zhou
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Yanjiao Wu
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Lifeng Ye
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Yunting Wang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Kaimin Zhang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Lingjun Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, China
| | - Yi Huang
- Department of Stomatology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Lei Wang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Shaoxiang Xian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, China
| | - Yang Zhang
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5037, USA
- Corresponding author. Tel.: +1 713 743 7710.
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
- Corresponding author. Tel.: +86 20 39357276.
| |
Collapse
|
16
|
Shen C, Liu J, Zhu F, Lei R, Cheng H, Zhang C, Sui X, Ding L, Yang M, Chen H, Ding R, Cao J. The effects of cooking oil fumes-derived PM 2.5 on blood vessel formation through ROS-mediated NLRP3 inflammasome pathway in human umbilical vein endothelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 174:690-698. [PMID: 30878009 DOI: 10.1016/j.ecoenv.2019.03.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/01/2019] [Accepted: 03/07/2019] [Indexed: 05/05/2023]
Abstract
BACKGROUND Cooking oil fumes (COFs), a main pollutant in kitchen air, is a major risk to human health. In our previous research, exposure to COFs-derived PM2.5 could cause umbilical vascular endothelial dysfunction, leading to decreased fetal weight. Here, to test the role of ROS-mediated NLRP3 inflammasome pathway in blood vessel formation of human umbilical vein endothelial cells (HUVECs) caused by COFs-derived PM2.5, the cells were exposed to COFs-derived PM2.5 at different concentrations with and without N-acetyl-L-cysteine (NAC). METHODS MTT assay was used to determine HUVECs viability. Intracellular ROS and mitochondrial ROS levels were assessed with DCFH-DA and MitoSOX™ assay. The levels of proteins and mRNA involved in NLRP3 inflammasome signaling pathway and VEGF were measured by western blot and real-time PCR (RT-PCR). Tube formation in HUVECs was detected by tube formation assay. RESULTS The results revealed that COFs-derived PM2.5 exposure reduced HUVECs viability, increased the intracellular and mitochondrial ROS levels in cells, and up-regulated the levels of proteins and mRNA involved in NLRP3 inflammasome signaling pathway. However, the protein and mRNA expression of VEGF were reduced with the increasing exposure concentrations. In addition, COFs-derived PM2.5 also affected the tube formation. However, co-incubation with NAC effectively rescued the damages caused by COFs-derived PM2.5 exposure. CONCLUSIONS This study proved that COFs-derived PM2.5 could significantly reduce HUVECs viability, induce the overproduction of ROS, lead to inflammation and inhibit VEGF expression, thus affect angiogenesis of HUVECs in vitro. It was revealed that the impact caused by COFs-derived PM2.5 on blood vessel formation through a ROS-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Chaowei Shen
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Jie Liu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Furong Zhu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Ruoqian Lei
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Han Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Chao Zhang
- Department of Environmental Health, Center for Disease Control and Prevention, Nanjing, China.
| | - Xinmiao Sui
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Liu Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Mei Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Hongbo Chen
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical Universit, 15 Yimin Road, Hefei, China.
| | - Rui Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Jiyu Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|
17
|
Hardeland R. Aging, Melatonin, and the Pro- and Anti-Inflammatory Networks. Int J Mol Sci 2019; 20:ijms20051223. [PMID: 30862067 PMCID: PMC6429360 DOI: 10.3390/ijms20051223] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Aging and various age-related diseases are associated with reductions in melatonin secretion, proinflammatory changes in the immune system, a deteriorating circadian system, and reductions in sirtuin-1 (SIRT1) activity. In non-tumor cells, several effects of melatonin are abolished by inhibiting SIRT1, indicating mediation by SIRT1. Melatonin is, in addition to its circadian and antioxidant roles, an immune stimulatory agent. However, it can act as either a pro- or anti-inflammatory regulator in a context-dependent way. Melatonin can stimulate the release of proinflammatory cytokines and other mediators, but also, under different conditions, it can suppress inflammation-promoting processes such as NO release, activation of cyclooxygenase-2, inflammasome NLRP3, gasdermin D, toll-like receptor-4 and mTOR signaling, and cytokine release by SASP (senescence-associated secretory phenotype), and amyloid-β toxicity. It also activates processes in an anti-inflammatory network, in which SIRT1 activation, upregulation of Nrf2 and downregulation of NF-κB, and release of the anti-inflammatory cytokines IL-4 and IL-10 are involved. A perhaps crucial action may be the promotion of macrophage or microglia polarization in favor of the anti-inflammatory phenotype M2. In addition, many factors of the pro- and anti-inflammatory networks are subject to regulation by microRNAs that either target mRNAs of the respective factors or upregulate them by targeting mRNAs of their inhibitor proteins.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, 37073 Göttingen, Germany.
| |
Collapse
|
18
|
Raut PK, Kim SH, Choi DY, Jeong GS, Park PH. Growth of breast cancer cells by leptin is mediated via activation of the inflammasome: Critical roles of estrogen receptor signaling and reactive oxygen species production. Biochem Pharmacol 2019; 161:73-88. [PMID: 30633869 DOI: 10.1016/j.bcp.2019.01.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022]
|
19
|
Chen X, Cao J, Sun Y, Dai Y, Zhu J, Zhang X, Zhao X, Wang L, Zhao T, Li Y, Liu Y, Wei G, Zhang T, Yan Z. Ethanol extract of Schisandrae chinensis fructus ameliorates the extent of experimentally induced atherosclerosis in rats by increasing antioxidant capacity and improving endothelial dysfunction. PHARMACEUTICAL BIOLOGY 2018; 56:612-619. [PMID: 31070526 PMCID: PMC6282463 DOI: 10.1080/13880209.2018.1523933] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
CONTEXT Schisandrae chinensis fructus, the dried ripe fruit of Schisandra chinensis (Turcz.) Baill. (Magnoliaceae) has been used for thousands of years as a traditional Chinese herb, which can attenuate and prevent the development of cardiovascular events. OBJECTIVE To evaluate the effects of the ethanol extracts from Schisandrae chinensis fructus fruit (EESC) on experimental atherosclerosis (AS) in rats. MATERIALS AND METHODS Treatment with EESC (0.35, 0.7, 1.4 g/kg/d, i.g.) and simvastatin (4 mg/kg/d, i.g.) on AS rats for 3 weeks. Sprague-Dawley rats on normal chow and under water treatment were used as control. The content of schisandrin, schisandrin A and schisandrin B in EESC was detected by HPLC. Aortic pathology changes, serum biochemical indices and nuclear factor E2-related factor 2 (Nrf-2) and heame oxygenase-1 (HO-1) expressions were measured. RESULTS Schisandrin, schisandrin A and schisandrin B contents were 291.8, 81.46 and 279.1 mg/g of dry weight, respectively. EESC significantly reduced the aortic plaque area (76.5, 90.5 and 73.9% reduction), regulated the levels of serum lipid (p < 0.05), enhanced the antioxidant enzyme activities (p < 0.01), reduced the malondialdehyde levels (72.5, 69.3, 67.3%), and up-regulated the Nrf-2 and HO-1 expression (p < 0.05). Furthermore, EESC reduced the levels of oxidized-LDL and endothelin-1 and thromboxane B2 but increased that of 6-keto prostaglandin F1α (p < 0.05). Acute toxicity was calculated on mice to be LD50 > 20 g/kg. CONCLUSIONS EESC positively affects the treatment of AS in vivo and the findings will provide a reliable theoretical basis for developing novel therapeutics.
Collapse
Affiliation(s)
- Xiu Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Jiahong Cao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Yong Sun
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Yaolan Dai
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Jiali Zhu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Xuemei Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Xiaoqin Zhao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Liwen Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Tingting Zhao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Yongbiao Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Youping Liu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Guihua Wei
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | - Tiane Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
- CONTACT Zhiyong Yan School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China; Tiane Zhang School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Zhiyong Yan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
- CONTACT Zhiyong Yan School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China; Tiane Zhang School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| |
Collapse
|
20
|
He Q, Li Z, Wang Y, Hou Y, Li L, Zhao J. Resveratrol alleviates cerebral ischemia/reperfusion injury in rats by inhibiting NLRP3 inflammasome activation through Sirt1-dependent autophagy induction. Int Immunopharmacol 2017; 50:208-215. [DOI: 10.1016/j.intimp.2017.06.029] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/06/2017] [Accepted: 06/26/2017] [Indexed: 01/07/2023]
|
21
|
Wang L, Chen K, Wan X, Wang F, Guo Z, Mo Z. NLRP3 inflammasome activation in mesenchymal stem cells inhibits osteogenic differentiation and enhances adipogenic differentiation. Biochem Biophys Res Commun 2017; 484:871-877. [PMID: 28167279 DOI: 10.1016/j.bbrc.2017.02.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 02/02/2017] [Indexed: 10/20/2022]
Abstract
Osteoporosis is one of the most common skeletal disease featured by osteopenia and adipose accumulation in bone tissue. NLRP3 inflammasome activation is an essential player in aging-related chronic diseases like osteoporosis, particularly due to the causal caspase-1 activation and its correlation to adipose accumulation in bone tissue. Moreover, the expression of anti-aging/senescence SIRT1 was reported to decline along with aging. As the major cellular contributor of bone formation, mesenchymal stem cells (MSCs) are multipotent stem cells processing mutually exclusive differentiatability toward osteocytes or adipocytes. Therefore, we hypothesized that NLRP3 inflammasome activation promotes adipogenesis and repress osteogenesis in MSCs via inhibiting SIRT1 expression. We activated NLRP3 inflammasome in human MSCs via lipopolysaccharide and palmitic acid (LPS/PA) treatment for self-renewal maintenance, adipogenic differentiation or osteogenic differentiation. LPS/PA treatment significantly increased NLRP3 expression, decreased SIRT1 expression and promoted caspase-1 activity in MSCs. LPS/PA treatment also boosted adipogenesis of MSCs and suppressed osteogenesis. Moreover, inhibition of caspase-1 activity repressed adipogenic differentiation and partially improved osteogenic differentiation of MSCs with LPS/PA treatment. Our study demonstrated the pivotal roles of NLRP3 inflammasome and downstream mediator caspase-1 for the progress of osteo-differentiation MSCs, and offered novel therapeutic target of treatment for osteoporosis.
Collapse
Affiliation(s)
- Linghao Wang
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Ke Chen
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Xinxing Wan
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Fang Wang
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Zi Guo
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Zhaohui Mo
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China.
| |
Collapse
|