1
|
Thangwong P, Tocharus C, Tocharus J. The Bidirectional Role of Hypoxia-Inducible Factor 1 Alpha in Vascular Dementia Caused by Chronic Cerebral Hypoperfusion. Mol Neurobiol 2025:10.1007/s12035-025-04914-5. [PMID: 40205304 DOI: 10.1007/s12035-025-04914-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Chronic cerebral hypoperfusion (CCH) is a critical indicator of cognitive impairment and dementia, especially vascular dementia. Cerebral blood flow disturbance alters the properties of neurons and glial cells as a result of a deficit in energy sources. Hypoxia-inducible factor 1 alpha (HIF- 1α) is a transcription factor that controls gene activity in response to low oxygen levels. It regulates a complex network of cellular adaptations to improve oxygenation, metabolic reprogramming, and cell survival in hypoxic situations. However, recent research suggests that HIF- 1α plays a role not only in neuroprotection but also in brain injury. It is therefore critical to fully comprehend the mechanisms behind these disorders. This review highlights the dual role of HIF- 1α in CCH-induced VaD. Initially, HIF- 1α provides a neuroprotection by promoting angiogenesis through vascular endothelial growth factor (VEGF) signaling. However, prolonged activation can detrimentally effects, including oxidative stress, neuroinflammation, blood-brain barrier dysfunction, and cognitive impairment. Evidence suggests that HIF- 1α exerts its protective effects in acute ischemic/hypoxic-induced VaD through pathways such as PI3 K/AKT/mTOR and MAPK/p-c-Jun signaling. However, its dysregulation in chronic stages of CCH contributes to cognitive decline and disease progression. Understanding the complex role of HIF- 1α and its interactions with other molecular pathways is crucial for developing effective therapeutic strategies. Therefore, an informed, in-depth discussion of its involvement in these pathologic processes is necessary, as a precise contribution of HIF- 1α to CCH-induced VaD remains to be established and requires further investigation.
Collapse
Affiliation(s)
- Phakkawat Thangwong
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Functional Food Research Center for Well-Being, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Functional Food Research Center for Well-Being, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
2
|
Lu C, Chen M, Zhao Y, Zhan Y, Wei X, Lu L, Yang M, Gong X. A Co-MOF encapsulated microneedle patch activates hypoxia induction factor-1 to pre-protect transplanted flaps from distal ischemic necrosis. Acta Biomater 2024; 184:171-185. [PMID: 38871202 DOI: 10.1016/j.actbio.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/25/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Avoiding ischemic necrosis after flap transplantation remains a significant clinical challenge. Developing an effective pretreatment method to promote flap survival postoperatively is crucial. Cobalt chloride (CoCl2) can increase cell tolerance to ischemia and hypoxia condition by stimulating hypoxia-inducible factor-1 (HIF-1) expression. However, the considerable toxic effects severely limit the clinical application of CoCl2. In this study, cobalt-based metal-organic frameworks (Co-MOF) encapsulated in a microneedle patch (Co-MOF@MN) was developed to facilitate the transdermal sustained release of Co2+ for rapid, minimally invasive rapid pretreatment of flap transplantation. The MN patch was composed of a fully methanol-based two-component cross-linked polymer formula, with a pyramid structure and high mechanical strength, which satisfied the purpose of penetrating the skin stratum corneum of rat back to achieve subcutaneous vascular area administration. Benefiting from the water-triggered disintegration of Co-MOF and the transdermal delivery via the MN patch, preoperative damage and side effects were effectively mitigated. Moreover, in both the oxygen-glucose deprivation/recovery (OGD/R) cell model and the rat dorsal perforator flap model, Co-MOF@MN activated the HIF-1α pathway and its associated downstream proteins, which reduced reperfusion oxidative damage, improved blood supply in choke areas, and increased flap survival rates post-transplantation. This preprotection strategy, combining MOF nanoparticles and the MN patch, meets the clinical demands for trauma minimization and uniform administration in flap transplantation. STATEMENT OF SIGNIFICANCE: Cobalt chloride (CoCl2) can stimulate the expression of hypoxia-inducible factor (HIF-1) and improve the tolerance of cells to ischemia and hypoxia conditions. However, the toxicity and narrow therapeutic window of CoCl2 severely limit its clinical application. Herein, we explored the role of Co-MOF as a biocompatible nanocage for sustained release of Co2+, showing the protective effect on vascular endothelial cells in the stress model of oxygen-glucose deprivation. To fit the clinical needs of minimal trauma in flap transplantation, a Co-MOF@MN system was developed to achieve local transdermal delivery at the choke area, significantly improving blood supply opening and flap survival rate. This strategy of two-step delivery of Co2+ realized the enhancement of biological functions while ensuring the biosafety.
Collapse
Affiliation(s)
- Cheng Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Miao Chen
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Yuanyuan Zhao
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Yongxin Zhan
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Xin Wei
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Laijin Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China; Orthopedics Central Laboratory, Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun 130021, PR China
| | - Mingxi Yang
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China; Orthopedics Central Laboratory, Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun 130021, PR China.
| | - Xu Gong
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China; Orthopedics Central Laboratory, Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
3
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
4
|
Xiao J, Zhou Y, Sun L, Wang H. Role of integrating cannabinoids and the endocannabinoid system in neonatal hypoxic-ischaemic encephalopathy. Front Mol Neurosci 2023; 16:1152167. [PMID: 37122621 PMCID: PMC10130673 DOI: 10.3389/fnmol.2023.1152167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/16/2023] [Indexed: 05/02/2023] Open
Abstract
Neonatal hypoxic-ischaemic events, which can result in long-term neurological impairments or even cell death, are among the most significant causes of brain injury during neurodevelopment. The complexity of neonatal hypoxic-ischaemic pathophysiology and cellular pathways make it difficult to treat brain damage; hence, the development of new neuroprotective medicines is of great interest. Recently, numerous neuroprotective medicines have been developed to treat brain injuries and improve long-term outcomes based on comprehensive knowledge of the mechanisms that underlie neuronal plasticity following hypoxic-ischaemic brain injury. In this context, understanding of the medicinal potential of cannabinoids and the endocannabinoid system has recently increased. The endocannabinoid system plays a vital neuromodulatory role in numerous brain regions, ensuring appropriate control of neuronal activity. Its natural neuroprotection against adult brain injury or acute brain injury also clearly demonstrate the role of endocannabinoid signalling in modulating neuronal activity in the adult brain. The goal of this review is to examine how cannabinoid-derived compounds can be used to treat neonatal hypoxic-ischaemic brain injury and to assess the critical function of the endocannabinoid system and its potential for use as a new neuroprotective treatment for neonatal hypoxic-ischaemic brain injury.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Pathology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Yue Zhou
- Department of Pharmacy, Xindu District People’s Hospital of Chengdu, Chengdu, China
| | - Luqiang Sun
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haichuan Wang
- Department of Paediatrics, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Haichuan Wang,
| |
Collapse
|
5
|
In Vitro Evaluation of the Shading Effect of Various Zirconia Surface Stains on Porcelain Crowns. COATINGS 2022. [DOI: 10.3390/coatings12060734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Human teeth display various colors under natural light. Dental restorations, such as zirconia crowns, are generally used to rehabilitate the oral function of patients with tooth loss due to trauma or natural tooth falls. However, significant improvements in the color and translucency of zirconia are required to meet the clinical needs for dental restoration. In the past, a large amount of ceramic powder has been used to improve the appearance of zirconia. However, the interface between the ceramic powder and zirconia makes them prone to falling off. Therefore, the aesthetics of zirconia crowns remains a major challenge. Recently, substantial advances have been made in the field of dental materials, as special staining agents for zirconia have been introduced as alternatives to ceramic powders. Therefore, this study tested zirconia-specific staining agents that were used to produce zirconia samples with A1 and A3 colors. A dental colorimetric plate was used as the control group to assess the staining effects of the different brands of staining agents. Meanwhile, two hypotheses were proposed: that the staining effects of these special staining agents for zirconia met the criteria for clinical application and that there was no significant difference between the different staining agents for zirconia. The results showed that the coatings of different brands of staining agents were ultrathin, with a thickness of approximately 27–78 µm. In addition, the coloring effects of the zirconia staining agents were not significantly different from those of the colorimetric plates. After staining, the zirconia samples had decreased surface roughness and contact angle values, which improved surface smoothness and cleanliness. In summary, the results support the hypothesis of this study that zirconia stains can be used as an alternative to the current fabrication methods for clinical dental restorations. We sought to identify the clinical techniques that are easier to perform and to overcome the current problem of dental technicians requiring considerable dentin space for staining. It is expected that the results of this study will be useful in clinical dental restorations.
Collapse
|
6
|
Wang W, Tang J, Zhong M, Chen J, Li T, Dai Y. HIF-1 α may play a role in late pregnancy hypoxia-induced autism-like behaviors in offspring rats. Behav Brain Res 2021; 411:113373. [PMID: 34048873 DOI: 10.1016/j.bbr.2021.113373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that can be caused by various factors. The present study aimed to determine whether prenatal hypoxia can lead to ASD and the role of hypoxia-inducible factor-1α (HIF-1α) in this process. We constructed a prenatal hypoxia model of pregnant rats by piping nitrogen and oxygen mixed gas, with an oxygen concentration of 10 ± 0.5 %, into the self-made hypoxia chamber. Rats were subjected to different extents of hypoxia treatments at different points during pregnancy. The results showed that hypoxia for 6 h on the 17th gestation day is most likely to lead to autistic behavior in offspring rats, including social deficits, repetitive behaviors, and impaired learning and memory. The mRNA expression level of TNF-α also increased in hypoxia-induced autism group and valproic acid (VPA) group. Western blotting analysis showed increased levels of hypoxia inducible factor 1 alpha (HIF-1α) and decreased levels of phosphatase and tensin homolog (PTEN) in the hypoxic-induced autism group. Meanwhile, N-methyl d-aspartate receptor subtype 2 (NR2A) and glutamate ionotropic receptor AMPA type subunit 2 (GluR2) were upregulated in the hypoxic-induced autism group. HIF-1α might play a role in hypoxia-caused autism-like behavior and its regulatory effect is likely to be achieved by regulating synaptic plasticity.
Collapse
Affiliation(s)
- Weiyu Wang
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jinghua Tang
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Min Zhong
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jie Chen
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Tingyu Li
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Ying Dai
- Department of Primary Child Health Care, Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, China; Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorder, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
| |
Collapse
|
7
|
Cardozo V, Vaamonde L, Parodi-Talice A, Zuluaga MJ, Agrati D, Portela M, Lima A, Blasina F, Dajas F, Bedó G. Multitarget neuroprotection by quercetin: Changes in gene expression in two perinatal asphyxia models. Neurochem Int 2021; 147:105064. [PMID: 33951501 DOI: 10.1016/j.neuint.2021.105064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/27/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) causes mortality and long-term neurologic morbidities in newborns, affecting pathways related to energy failure, excitotoxicity and oxidative stress that often lead to cell death. The whole process of HIE injury is coupled to changes in the expression of a great array of proteins. A nanoliposomal preparation of the flavonoid quercetin has been shown to exert neuroprotective effects in perinatal asphyxia models. This study aimed to identify neonatal HIE markers and explore the effect of quercetin administration in two perinatal asphyxia models: newborn rats and piglets. In the rat model, nanoliposomal quercetin administration reduced mortality after asphyxia. In the piglet model, quercetin partially overrode the reduction of HIF-1α mRNA levels in the cortex induced by asphyxia. Quercetin administration also reduced increased level of HO-1 mRNA in asphyctic piglets. These results suggest that quercetin neuroprotection might be involved in the regulation of HIF-1α, HO-1 and their targets. A proteomic approach revealed that the glycolytic pathway is strongly regulated by quercetin in both species. We also identified a set of proteins differentially expressed that could be further considered as markers. In piglets, this set includes Acidic Leucine-rich nuclear phosphoprotein 32 (ANP32A), associated with nervous system differentiation, proteins related with death pathways and alpha-enolase which can be converted to neuron-specific enolase, a glycolytic enzyme that may promote neuroprotection. In newborn rats, other promising proteins associated with neurogenesis and neuroprotection emerged, such as dihydropyrimidinase-related proteins, catalytic and regulatory subunits of phosphatases and heterogeneous nuclear ribonucleoprotein K (hnRNPK). Our results show that a nanoliposomal preparation of quercetin, with protective effect in two HIE mammal models, modulates the expression of proteins involved in energy metabolism and other putative neuroprotective signals in the cortex. Identification of these signals could reveal potential molecular pathways involved in disease onset and the novel quercetin neuroprotective strategy.
Collapse
Affiliation(s)
- V Cardozo
- Sección Genética Evolutiva, Facultad de Ciencias, Universidad de la República (Udelar), Montevideo, Uruguay
| | - L Vaamonde
- Dept. Neonatología, Facultad de Medicina, Universidad de la República (Udelar), Montevideo, Uruguay
| | - A Parodi-Talice
- Sección Genética Evolutiva, Facultad de Ciencias, Universidad de la República (Udelar), Montevideo, Uruguay; Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - M J Zuluaga
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República (Udelar), Montevideo, Uruguay
| | - D Agrati
- Sección Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República (Udelar), Montevideo, Uruguay
| | - M Portela
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo; Facultad de Ciencias, Universidad de la República (Udelar), Montevideo, Uruguay
| | - A Lima
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo; Instituto de Investigaciones Biológicas Clemente Estable, Ministerio de Educación y Cultura, Montevideo, Uruguay
| | - F Blasina
- Dept. Neonatología, Facultad de Medicina, Universidad de la República (Udelar), Montevideo, Uruguay.
| | - F Dajas
- Dept. Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, Ministerio de Educación y Cultura, Montevideo, Uruguay
| | - G Bedó
- Sección Genética Evolutiva, Facultad de Ciencias, Universidad de la República (Udelar), Montevideo, Uruguay.
| |
Collapse
|
8
|
Kukec E, Goričar K, Dolžan V, Rener-Primec Z. HIF1A polymorphisms do not modify the risk of epilepsy nor cerebral palsy after neonatal hypoxic-ischemic encephalopathy. Brain Res 2021; 1757:147281. [PMID: 33515534 DOI: 10.1016/j.brainres.2021.147281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 11/19/2022]
Abstract
PURPOSE Hypoxic-ischemic encephalopathy (HIE) remains the major cause of cerebral palsy and epilepsy in developed countries. Hypoxia-inducible factor 1 alpha (HIF-1α) is the key mediator of oxygen homoeostasis. The aim of this study was to investigate whether hypoxia-inducible factor 1 subunit alpha (HIF1A) functional polymorphisms are associated with the risk of epilepsy, drug-resistant epilepsy, and cerebral palsy after neonatal HIE. METHODS The study included 139 healthy controls and 229 patients with epilepsy and/or cerebral palsy, of which 95 had perinatal HIE. Genomic DNA isolated from buccal swabs or peripheral blood were genotyped for HIF1A rs11549465 and rs11549467 using PCR based methods. RESULTS The investigated HIF1A polymorphisms did not influence the risk of epilepsy and its drug-resistance nor cerebral palsy after neonatal HIE (all p > 0.05). Clinical characteristics of patients were significantly associated with neurological deficits after HIE. CONCLUSION This study found no statistically significant association of HIF1A rs11549465 and rs11549467 with the development of epilepsy and its drug-resistance, as well as cerebral palsy, after neonatal HIE.
Collapse
Affiliation(s)
- Eva Kukec
- Department of Child, Adolescent, and Developmental Neurology, Children's Hospital, University Medical Centre Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia
| | - Katja Goričar
- Faculty of Medicine, University of Ljubljana, Slovenia; Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Vita Dolžan
- Faculty of Medicine, University of Ljubljana, Slovenia; Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Zvonka Rener-Primec
- Department of Child, Adolescent, and Developmental Neurology, Children's Hospital, University Medical Centre Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia.
| |
Collapse
|
9
|
Lin WC, Yao C, Huang TY, Cheng SJ, Tang CM. Long-term in vitro degradation behavior and biocompatibility of polycaprolactone/cobalt-substituted hydroxyapatite composite for bone tissue engineering. Dent Mater 2019; 35:751-762. [PMID: 30857736 DOI: 10.1016/j.dental.2019.02.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Currently, infections due to foreign-body reactions caused by bacteria or implant materials at the wound site are one of the major reasons for the failure of guided tissue regeneration (GTR) and guided bone regeneration (GBR) in clinical applications. The purpose of this study was to develop regeneration membranes with localized cobalt ion release to reduce infection and inflammation by polycaprolactone (PCL)/cobalt-substituted hydroxyapatite (CoHA). METHODS The PCL composite membrane containing 20 wt% CoHA powders was prepared by solvent casting. The surface morphology, crystal structure, chemical composition and thermal properties of PCL composite membranes were characterized. The biocompatibility, osteogenic differentiation and antibacterial properties of composite membrane were also investigated. Then, in biodegradability was assessed by immersing phosphate buffer solution (PBS) for 6 months. RESULTS Physicochemical analyses revealed that CoHA is evenly mixed in the membranes and assistance reduce the crystallinity of PCL for getting more degradation amounts than PCL membrane. Osteoblast cells culture on the membrane showed that the CoHA significantly increases cell proliferation and found the calcium deposition production increased over 90% compared with PCL after 7 days of culture. A good antibacterial effect was achieved by the addition of CoHA powder. The results were confirmed by 2.4 times reduction of proliferation of Escherichia coli (E. coli) seeded on the composite membrane after 24 h. Immersing in PBS for 6 months indicated that PCL-CoHA composite membrane has improved biodegradation and can continuously remove free radicals to reduce the inflammatory response. SIGNIFICANCE The PCL-CoHA composite membrane with suitable releasing of cobalt ion can be considered as a potential choice for bone tissue regeneration.
Collapse
Affiliation(s)
- Wei-Chun Lin
- Graduate Institute of Oral Science, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Chenmin Yao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Ting-Yun Huang
- Graduate Institute of Oral Science, Chung Shan Medical University, Taichung 40201, Taiwan.
| | - Shih-Jung Cheng
- Department of Dentistry, Chung Shan Medical University, Taiwan.
| | - Cheng-Ming Tang
- Graduate Institute of Oral Science, Chung Shan Medical University, Taichung 40201, Taiwan; Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
| |
Collapse
|
10
|
Yang X, Zhong M, Chen J, Li T, Cheng Q, Dai Y. HIF-1<alpha> Repression of PTEN Transcription Mediates Protective Effects of BMSCs on Neurons During Hypoxia. Neuroscience 2018; 392:57-65. [PMID: 30267829 DOI: 10.1016/j.neuroscience.2018.09.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/06/2018] [Accepted: 09/18/2018] [Indexed: 01/14/2023]
Abstract
Neonatal hypoxic-ischemic brain damage (HIBD) is a cerebral hypoxic-ischemic disease caused by a variety of insults during the perinatal period, leading to varying degrees of cognitive dysfunction. Mesenchymal stem cells play an important role in functional recovery, but the mechanism is not yet clear. It has been reported that HIF-1<alpha> and PTEN are involved in the process of hypoxia-ischemia, but the specific roles that these proteins play remains to be understood. In this study, we performed oxygen glucose deprivation (OGD) or CoCl2 preconditioning on hippocampal neurons to simulate a hypoxic environment in vitro, and then co-cultured them with BMSCs, to observe the effect of BMSCs and the role of HIF-1<alpha>. In addition, bpV, an inhibitor of PTEN was added to OGD neurons to determine the role of PTEN during hypoxia. We found that the levels of cell damage and apoptosis in OGD neurons decreased significantly after co-culture with BMSCs. Apoptosis was increased when HIF-1<alpha> was inhibited, but neurons remained protected when PTEN was suppressed. We further established that HIF-1<alpha> was enriched at the PTEN promoter both in BMSCs and hippocampal neurons, with increased enrichment under hypoxic conditions, leading to reduced transcription of PTEN. Our findings support the conclusion that CoCl2 preconditioning of BMSCs can simulate hypoxic conditions and can protect OGD neurons, an effect that is mediated through activation of the HIF-1<alpha> system and repression of PTEN transcription.
Collapse
Affiliation(s)
- Xin Yang
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Min Zhong
- Department of Neurology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jie Chen
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Tingyu Li
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Qian Cheng
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
| | - Ying Dai
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
| |
Collapse
|
11
|
Cao S, Du J, Hei Q. Lycium barbarum polysaccharide protects against neurotoxicity via the Nrf2-HO-1 pathway. Exp Ther Med 2017; 14:4919-4927. [PMID: 29201196 PMCID: PMC5704330 DOI: 10.3892/etm.2017.5127] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 06/08/2017] [Indexed: 12/15/2022] Open
Abstract
The incidence of neurodegenerative diseases including Alzheimer's and Parkinson's disease has markedly increased over the past few decades. Oxidative stress is considered to be a common pathophysiological condition resulting in neurotoxicity. Lycium barbarum polysaccharide (LBP) is the major active component of Lycium barbarum L., which exhibit potent antioxidant activity. The current study investigated the neuroprotective effects of LBP in H2O2-treated PC12 cells in vitro and in CoCl2-treated rats in vivo. It was determined that LBP concentration-dependently reversed the H2O2-induced increase in reactive oxygen species (ROS) levels, decrease in cell viability, increase in TUNEL-stained cells, increase in caspase-3 and −9 activity and decrease in mitochondrial membrane potential, indicating the amelioration of mitochondrial apoptosis. Furthermore, LBP inhibited the H2O2-induced decrease in nuclear factor erythroid 2-related factor 2 (Nrf)2 and heme oxygenase (HO)-1 expression and binding of Nrf2 to the promoters of HO-1. Silencing of Nrf2 and inhibition of HO-1 by zinc protoporphyrin IX (ZnPP) reversed the protective effects of LBP against H2O2-resulted neurotoxicity in PC12 cells. In CoCl2-treated rats, it was demonstrated that LBP decreased brain tissue apoptosis, reduced the time spent by rats finding the platform site, decreased escape latencies and reduced the distance traveled to find the platform. In addition, LBP inhibited the CoCl2-induced decrease of Nrf2 and HO-1 expression. Administration of ZnPP also suppressed the protective effects of LBP against CoCl2-resulted neurotoxicity in rats. Thus, the current study indicated that LBP exhibits protective effects against neurotoxicity by upregulating Nrf2/HO-1 signaling. These data may increase understanding regarding the neuroprotective activities of LBP.
Collapse
Affiliation(s)
- Shumei Cao
- Department of Anesthesiology, Xi'an No. 1 Hospital, Xi'an, Shaanxi 710002, P.R. China
| | - Jianlong Du
- Department of Anesthesiology, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi 712000, P.R. China
| | - Qiaohong Hei
- Department of Anesthesiology, Xi'an High-Tech Hospital, Xi'an, Shaanxi 710075, P.R. China
| |
Collapse
|
12
|
Dai Y, Li W, Zhong M, Chen J, Cheng Q, Liu Y, Li T. The paracrine effect of cobalt chloride on BMSCs during cognitive function rescue in the HIBD rat. Behav Brain Res 2017; 332:99-109. [PMID: 28576310 DOI: 10.1016/j.bbr.2017.05.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/20/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022]
Abstract
Hypoxia-ischemia (HI)-induced perinatal encephalopathy frequently causes chronic neurological morbidities and acute mortality. Bone mesenchymal stem cell (BMSC) transplantation could potentially promote functional and anatomical recovery of ischemic tissue. In vitro hypoxic preconditioning is an effective strategy to improve the survival of BMSCs in ischemic tissue. In this study, cobalt chloride (CoCl2) preconditioned medium from BMSC cultures was injected into the left lateral ventricle of HI rats using a micro-osmotic pump at a flow rate 1.0μl/h for 7 days. The protein levels of HIF-1α and its target genes, vascular endothelial growth factor and erythropoietin, markedly increased after CoCl2 preconditioning in BMSCs. In 7-week-old rats that received CoCl2 preconditioned BMSC medium, results of the Morris water maze test indicated ameliorated spatial working memory function following hypoxia-ischemia damage. Neuronal loss, cellular disorganization, and shrinkage in brain tissue were also ameliorated. Extracellular field excitatory postsynaptic potentials (fEPSPs) in the brain slices of 8-week-old rats were recorded; administration of CoCl2 preconditioned BMSC culture medium induced a progressive increment of baseline and amplitude of the fEPSPs. Immunohistochemical quantification showed that GluR2 protein expression increased. In conclusion, CoCl2 activates HIF-1α signals in BMSCs. CoCl2 preconditioned BMSC culture medium likely effects neuroprotection by inducing long-term potentiation (LTP), which could be associated with GluR2 expression. The paracrine effects of hypoxia preconditioning on BMSCs could have applications in novel cell-based therapeutic strategies for hypoxic and ischemic brain injury.
Collapse
MESH Headings
- Animals
- Brain/pathology
- Brain/physiopathology
- Cells, Cultured
- Cobalt/pharmacology
- Culture Media, Conditioned
- Disease Models, Animal
- Excitatory Postsynaptic Potentials
- Female
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Ischemia, Brain/pathology
- Hypoxia-Ischemia, Brain/physiopathology
- Hypoxia-Ischemia, Brain/psychology
- Hypoxia-Ischemia, Brain/therapy
- Male
- Maze Learning/physiology
- Memory, Short-Term/physiology
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/physiology
- Neuroprotection/physiology
- Protective Agents/pharmacology
- Rats, Sprague-Dawley
- Receptors, AMPA/metabolism
- Spatial Memory/physiology
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Ying Dai
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Wendi Li
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Min Zhong
- Department of Neurology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jie Chen
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China
| | - Qian Cheng
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Youxue Liu
- Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| | - Tingyu Li
- Department of Primary Child Health Care, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China; Pediatric Research Institute, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, China.
| |
Collapse
|
13
|
Zhao B, Zheng Z. Insulin Growth Factor 1 Protects Neural Stem Cells Against Apoptosis Induced by Hypoxia Through Akt/Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase (Akt/MAPK/ERK) Pathway in Hypoxia-Ishchemic Encephalopathy. Med Sci Monit 2017; 23:1872-1879. [PMID: 28420864 PMCID: PMC5405785 DOI: 10.12659/msm.901055] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Insulin growth factor 1 (IGF-1) is reported to modulate cell growth and acts as potential therapy for traumatic brain injury. This study was designed to investigate the effect of IGF-1 on hypoxia-induced apoptosis in neural stem cells (NSCs). Material/Methods A hypoxia model was constructed using NSCs separated from the hippocampus of rat. NSCs were divided into four groups: cells under normoxic conditions that acted as controls (C group), cells under hypoxia (H group), cells under hypoxia with IGF-1 (HI group), and cells under hypoxia with IGF-1 as well as picropodophyllin (PPP), which acts as an inhibitor of the IGF-1 receptor (HIP group). The cell viability and apoptosis were respectively measured by 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and flow cytometry. Finally, the phosphorylation levels of apoptosis-associated proteins and key kinases in the phosphatidylinositol-3-kinase (PI3K)/AKT and the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways were detected by Western blot analysis. Results In comparison with the H group, the cell viability was increased while the cell apoptosis was reduced by IGF-1 in the HI group. Besides, the expression levels of Bax, cytochrome c, and activated caspase-3 were all improved in the H group, and the remarkable differences were eliminated in the HI group compared with the C group. The expression level of Bcl-2 was the opposite. Additionally, down-regulations of phosphorylated AKT, MAPK, and ERK induced by hypoxia were all improved by IGF-1. All the influences of IGF-1 were weakened by addition of PPP. Conclusions IGF-1 increased cell viability while decreasing apoptosis in hypoxic NSCs through the PI3K/AKT and MAPK/ERK pathways.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Children Rehabilitation, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Zebao Zheng
- Department of Children Rehabilitation, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
14
|
Abstract
Stroke is the second most common cause of death and the leading cause of disability worldwide. Brain injury following stroke results from a complex series of pathophysiological events including excitotoxicity, oxidative and nitrative stress, inflammation, and apoptosis. Moreover, there is a mechanistic link between brain ischemia, innate and adaptive immune cells, intracranial atherosclerosis, and also the gut microbiota in modifying the cerebral responses to ischemic insult. There are very few treatments for stroke injuries, partly owing to an incomplete understanding of the diverse cellular and molecular changes that occur following ischemic stroke and that are responsible for neuronal death. Experimental discoveries have begun to define the cellular and molecular mechanisms involved in stroke injury, leading to the development of numerous agents that target various injury pathways. In the present article, we review the underlying pathophysiology of ischemic stroke and reveal the intertwined pathways that are promising therapeutic targets.
Collapse
|
15
|
Li L, Qu Y, Jin X, Guo XQ, Wang Y, Qi L, Yang J, Zhang P, Li LZ. Protective effect of salidroside against bone loss via hypoxia-inducible factor-1α pathway-induced angiogenesis. Sci Rep 2016; 6:32131. [PMID: 27558909 PMCID: PMC4997314 DOI: 10.1038/srep32131] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 08/03/2016] [Indexed: 12/02/2022] Open
Abstract
Hypoxia-inducible factor (HIF)-1α plays a critical role in coupling angiogenesis with osteogenesis during bone development and regeneration. Salidroside (SAL) has shown anti-hypoxic effects in vitro and in vivo. However, the possible roles of SAL in the prevention of hypoxia-induced osteoporosis have remained unknown. Two osteoblast cell lines, MG-63 and ROB, were employed to evaluate the effects of SAL on cell viability, apoptosis, differentiation and mineralization in vitro. Rats subjected to ovariectomy-induced bone loss were treated with SAL in vivo. Our results showed that pre-treatment with SAL markedly attenuated the hypoxia-induced reductions in cell viability, apoptosis, differentiation and mineralization. SAL down-regulated HIF-1α expression and inhibited its translocation; however, SAL increased its transcriptional activity and, consequently, up-regulated vascular endothelial growth factor (VEGF). In vivo studies further demonstrated that SAL caused decreases in the mineral, alkaline phosphatase (ALP), and BGP concentrations in the blood of ovariectomized (OVX) rats. Moreover, SAL improved the trabecular bone microarchitecture and increased bone mineral density in the distal femur. Additionally, SAL administration partially ameliorated this hypoxia via the HIF-1α-VEGF signalling pathway. Our results indicate that SAL prevents bone loss by enhancing angiogenesis and osteogenesis and that these effects are associated with the activation of HIF-1α signalling.
Collapse
Affiliation(s)
- Ling Li
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, People's Republic of China.,Department of Pharmacology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Ye Qu
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Xin Jin
- Department of Pharmacology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Xiao Qin Guo
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Yue Wang
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, People's Republic of China.,Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Lin Qi
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Jing Yang
- Department of Pathogenic Biology and Immunology, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Peng Zhang
- Department of Orthopaedics, Affiliated Hospital of Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| | - Ling Zhi Li
- Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard, Tianjin, People's Republic of China.,Department of Pharmaceutical Chemistry, Logistics College of Chinese People's Armed Police Forces, Tianjin, People's Republic of China
| |
Collapse
|
16
|
Beraudi A, Bianconi E, Catalani S, Canaider S, De Pasquale D, Apostoli P, Bordini B, Stea S, Toni A, Facchin F. In vivo response of heme-oxygenase-1 to metal ions released from metal-on-metal hip prostheses. Mol Med Rep 2016; 14:474-80. [PMID: 27176599 DOI: 10.3892/mmr.2016.5245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 01/20/2016] [Indexed: 11/06/2022] Open
Abstract
Metal ion release and accumulation is considered to be a factor responsible for the high failure rates of metal-on-metal (MoM) hip implants. Numerous studies have associated the presence of these ions, besides other factors, including a hypoxia‑like response and changes in pH due to metal corrosion leading to the induction of the oxidative stress response. The aim of the present study was to verify whether, in patients with a MoM hip prosthesis, mRNA and protein expression of HMOX‑1 was modulated by the presence of metal ions and whether patients without prostheses exhibit a different expression pattern of this enzyme. The study was conducted on 22 matched pairs of patients with and without prostheses, for a total of 44 samples. Ion dosage was determined using inductively coupled plasma mass spectrometry equipped with dynamic cell reaction. HMOX‑1 gene expression was quantified by reverse transcription-quantitative polymerase chain reaction and HMOX‑1 protein expression was analyzed using an enzyme-linked immunosorbent assay. The results demonstrated that although there were significant differences in the metallic ion concentrations amongst the two groups of patients, there was no correlation between circulating levels of cobalt (Co) and chromium (Cr), and HMOX‑1 gene and protein expression. Additionally, there was no significant difference in the protein expression levels of HMOX‑1 between the two groups. In conclusion, it was demonstrated that circulating Co and Cr ions released by articular prosthetics do not induce an increase in HMOX‑1 mRNA and protein expression at least 3.5 years after the implant insertion. The present study suggests that involvement of HMOX‑1 may be excluded from future studies and suggests that other antioxidant enzymes, including superoxide dismutase, glutathione peroxidase and reductase should be investigated.
Collapse
Affiliation(s)
- Alina Beraudi
- Medical Technology Laboratory, Rizzoli RIT Department ‑ Research, Innovation and Technology, Rizzoli Orthopaedic Institute, I‑40136 Bologna, Italy
| | - Eva Bianconi
- Department of Experimental, Diagnostic and Specialty Medicine, (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, I‑40136 Bologna, Italy
| | - Simona Catalani
- Unit of Occupational Health and Industrial Hygiene, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, I‑25100 Brescia, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine, (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, I‑40136 Bologna, Italy
| | - Dalila De Pasquale
- Medical Technology Laboratory, Rizzoli RIT Department ‑ Research, Innovation and Technology, Rizzoli Orthopaedic Institute, I‑40136 Bologna, Italy
| | - Pietro Apostoli
- Unit of Occupational Health and Industrial Hygiene, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, I‑25100 Brescia, Italy
| | - Barbara Bordini
- Medical Technology Laboratory, Rizzoli RIT Department ‑ Research, Innovation and Technology, Rizzoli Orthopaedic Institute, I‑40136 Bologna, Italy
| | - Susanna Stea
- Medical Technology Laboratory, Rizzoli RIT Department ‑ Research, Innovation and Technology, Rizzoli Orthopaedic Institute, I‑40136 Bologna, Italy
| | - Aldo Toni
- Medical Technology Laboratory, Rizzoli RIT Department ‑ Research, Innovation and Technology, Rizzoli Orthopaedic Institute, I‑40136 Bologna, Italy
| | - Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine, (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, I‑40136 Bologna, Italy
| |
Collapse
|
17
|
Guan D, Su Y, Li Y, Wu C, Meng Y, Peng X, Cui Y. Tetramethylpyrazine inhibits CoCl2
-induced neurotoxicity through enhancement of Nrf2/GCLc/GSH and suppression of HIF1α/NOX2/ROS pathways. J Neurochem 2015; 134:551-65. [DOI: 10.1111/jnc.13161] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/07/2015] [Accepted: 04/27/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Dongsheng Guan
- Department of the Second Clinical Medical College; Henan University of Traditional Chinese Medicine; Zhengzhou China
| | - Yufei Su
- Department of Emergency; Children's Hospital of Xi'an; Xi'an China
| | - Yingxia Li
- Department of the Second Clinical Medical College; Henan University of Traditional Chinese Medicine; Zhengzhou China
| | - Chuanjie Wu
- Department of Neurology; The First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Yi Meng
- Department of the Second Clinical Medical College; Henan University of Traditional Chinese Medicine; Zhengzhou China
| | - Xin Peng
- Department of the Second Clinical Medical College; Henan University of Traditional Chinese Medicine; Zhengzhou China
| | - Yinglin Cui
- Department of the Second Clinical Medical College; Henan University of Traditional Chinese Medicine; Zhengzhou China
| |
Collapse
|
18
|
Yan L, Cai Q, Xu Y. Hypoxic conditions differentially regulate TAZ and YAP in cancer cells. Arch Biochem Biophys 2014; 562:31-6. [PMID: 25078107 DOI: 10.1016/j.abb.2014.07.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 07/18/2014] [Accepted: 07/19/2014] [Indexed: 10/25/2022]
Abstract
The Hippo-YAP pathway is altered and implicated as an oncogenic signaling pathway in many human cancers. Hypoxia is an important microenvironmental factor that promotes tumorigenesis. However, the effects of hypoxia on the two most important Hippo-YAP effectors, YAP (Yes-associated protein) and TAZ (transcriptional co-activator with PDZ-binding motif), have not been reported. In this work, we demonstrated that TAZ was functionally involved in cell proliferation and/or migration in epithelial ovarian cancer (EOC) or human ovarian surface epithelial (HOSE) cells. Hypoxic conditions (1% O2 or hypoxia mimics) induced a reduction of YAP phosphorylation (S127) and total YAP expression in EOC cell lines OVCAR5 and SKOV3. However, these conditions up-regulated levels of S69 phosphorylated TAZ in EOC cells. The known TAZ kinases, Lats1 and Akt, were unlikely to be involved in up-regulated pTAZ by hypoxic conditions. Together, our data revealed new and differential regulating mechanisms of TAZ and YAP in cancer cells by hypoxia conditions.
Collapse
Affiliation(s)
- Libo Yan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 975 W. Walnut St. IB355A, Indianapolis, IN 46202, United States
| | - Qingchun Cai
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 975 W. Walnut St. IB355A, Indianapolis, IN 46202, United States
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 975 W. Walnut St. IB355A, Indianapolis, IN 46202, United States.
| |
Collapse
|