103 results on '"Qingyi Ma"'
Search Results
2. Correction: Blood–brain barrier-associated pericytes internalize and clear aggregated amyloid-β42 by LRP1-dependent apolipoprotein E isoform-specific mechanism
- Author
-
Qingyi Ma, Zhen Zhao, Abhay P. Sagare, Yingxi Wu, Min Wang, Nelly Chuqui Owens, Philip B. Verghese, Joachim Herz, David M. Holtzman, and Berislav V. Zlokovic
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Published
- 2024
- Full Text
- View/download PDF
3. Chronic kidney disease in a giant panda (Ailuropoda melanoleuca): a case report
- Author
-
Lingling Chang, Xiangyang L. Wang, Chenfei Yu, Chen-Hsuan Liu, Qiang Zhang, Yaping Wu, Ruoyi Jia, Qingyi Ma, Guanglin Pan, Dewen Tong, and Xinglong Wang
- Subjects
Giant panda ,Chronic kidney disease ,Heart failure ,Hypertension ,Veterinary medicine ,SF600-1100 - Abstract
Abstract Background Chronic kidney disease (CKD) is a common cause of morbidity and mortality in captive wildlife species. However, CKD has been rarely documented in giant pandas. Case presentation The following report describes a case of an eight-year-old female giant panda showing clinical signs of epistaxis, bloody diarrhea, polyuria, azotemia and anemia. The animal died despite of supportive treatments. Necropsy was performed. Grossly, both kidneys were shrunken and scarred with pallor. Subcutis edema and petechia on the epicardium of the heart were observed. The tissue samples were made into paraffin sections and stained by H.E and special staining including Periodic Acid-Schiff (PAS), von Kossa, Masson’s trichrome, Phosphotungstic acid-hematoxylin (PTAH), and Congo red. Histopathology examination revealed severe chronic tubulointerstitial nephritis with marked interstitial fibrosis, glomerulosclerosis, tubular atrophy and calcification in kidneys, and acute necrotizing hemorrhagic myocarditis with calcification in heart. Other lesions included intestinal hemorrhage, hepatic fatty degeneration and necrosis with hemosiderin, and splenic hemosiderin. Conclusions In summary, chronic kidney disease was finally diagnosed based on the association of clinical, gross, and histopathological findings. Heart failure secondary to CKD is the leading cause of death in this giant panda. The potential cause of CKD in this animal is possibly due to long term and uncontrolled hypertension. Blood pressure monitoring is essential in establishing the diagnosis and management of hypertension in giant panda.
- Published
- 2023
- Full Text
- View/download PDF
4. Avibacterium paragallinarum: an emerging birds pathogen in Qinling wildlife conservation center, China
- Author
-
Honglin Xie, Hui Li, Chenfei Yu, Yongqiang Miao, Yaping Wu, Ruoyi Jia, Qiang Zhang, Guanglin Pan, Qingyi Ma, Kangsheng Jia, and Xinglong Wang
- Subjects
Infectious coryza ,Avibacterium paragallinarum ,Avian diseases ,Pathogen ,Wlidlife ,Case report ,Veterinary medicine ,SF600-1100 ,Public aspects of medicine ,RA1-1270 - Abstract
Abstract The bacterium Avibacterium paragallinarum, previously known as Haemophilus paragallinarum, is responsible for causing infectious coryza (IC) in chickens and other avian species. In this case report, an outbreak of Avibacterium paragallinarum occurred in the Qinling area of China, resulting in clinical symptoms of facial swelling in several bird species, including Golden pheasant, Temminck's tragopan, and Peafowls, and three Golden pheasants died due to prolonged infection. Specific PCR results confirmed the presence of the pathogen in the infected birds. The report describes the clinical symptoms and pathological changes observed in the affected birds, as well as the isolation and identification of Avibacterium paragallinarum. Whole-genome sequencing and phylogenetic analysis were performed, and this is the first report of inter- and intra-species transmission of infectious coryza among wild birds in China.
- Published
- 2023
- Full Text
- View/download PDF
5. Author Correction: Pericyte loss influences Alzheimer-like neurodegeneration in mice
- Author
-
Abhay P. Sagare, Robert D. Bell, Zhen Zhao, Qingyi Ma, Ethan A. Winkler, Anita Ramanathan, and Berislav V. Zlokovic
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
6. Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
- Author
-
Isaac Kremsky, Qingyi Ma, Bo Li, Chiranjib Dasgupta, Xin Chen, Samir Ali, Shawnee Angeloni, Charles Wang, and Lubo Zhang
- Subjects
Fetal hypoxia ,Rat brain RNA-seq ,NF kappa B ,Biotechnology ,TP248.13-248.65 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Fetal hypoxia causes vital, systemic, developmental malformations in the fetus, particularly in the brain, and increases the risk of diseases in later life. We previously demonstrated that fetal hypoxia exposure increases the susceptibility of the neonatal brain to hypoxic-ischemic insult. Herein, we investigate the effect of fetal hypoxia on programming of cell-specific transcriptomes in the brain of neonatal rats. Results We obtained RNA sequencing (RNA-seq) data from neurons, microglia, oligodendrocytes, A2B5+ oligodendrocyte precursor cells, and astrocytes from male and female neonatal rats subjected either to fetal hypoxia or control conditions. Substantial transcriptomic responses to fetal hypoxia occurred in neurons, microglia, oligodendrocytes, and A2B5+ cells. Not only were the transcriptomic responses unique to each cell type, but they also occurred with a great deal of sexual dimorphism. We validated differential expression of several genes related to inflammation and cell death by Real-time Quantitative Polymerase Chain Reaction (qRT-PCR). Pathway and transcription factor motif analyses suggested that the NF-kappa B (NFκB) signaling pathway was enriched in the neonatal male brain due to fetal hypoxia, and we verified this result by transcription factor assay of NFκB-p65 in whole brain. Conclusions Our study reveals a significant impact of fetal hypoxia on the transcriptomes of neonatal brains in a cell-specific and sex-dependent manner, and provides mechanistic insights that may help explain the development of hypoxic-ischemic sensitive phenotypes in the neonatal brain.
- Published
- 2023
- Full Text
- View/download PDF
7. Retraction Note: Pericyte loss influences Alzheimer-like neurodegeneration in mice
- Author
-
Abhay P. Sagare, Robert D. Bell, Zhen Zhao, Qingyi Ma, Ethan A. Winkler, Anita Ramanathan, and Berislav V. Zlokovic
- Subjects
Science - Published
- 2024
- Full Text
- View/download PDF
8. Correction: Chronic kidney disease in a giant panda (Ailuropoda melanoleuca): a case report
- Author
-
Lingling Chang, Xiangyang L. Wang, Chenfei Yu, Chen-Hsuan Liu, Qiang Zhang, Yaping Wu, Ruoyi Jia, Qingyi Ma, Guanglin Pan, Dewen Tong, and Xinglong Wang
- Subjects
Veterinary medicine ,SF600-1100 - Published
- 2023
- Full Text
- View/download PDF
9. 3D doppler ultrasound imaging of cerebral blood flow for assessment of neonatal hypoxic-ischemic brain injury in mice.
- Author
-
Guofang Shen, Kayla Sanchez, Shirley Hu, Zhen Zhao, Lubo Zhang, and Qingyi Ma
- Subjects
Medicine ,Science - Abstract
Cerebral blood flow (CBF) acutely reduces in neonatal hypoxic-ischemic encephalopathy (HIE). Clinic studies have reported that severe CBF impairment can predict HIE outcomes in neonates. Herein, the present study uses a non-invasive 3D ultrasound imaging approach to evaluate the changes of CBF after HI insult, and explores the correlation between CBF alterations and HI-induced brain infarct in mouse pups. The neonatal HI brain injury was induced in postnatal day 7 mouse pups using the Rice-Vannucci model. Non-invasive 3D ultrasound imaging was conducted to image CBF changes with multiple frequencies on mouse pups before common carotid artery (CCA) ligation, immediately after ligation, and 0 or 24 hours after HI. Vascularity ratio of the ipsilateral hemisphere was acutely reduced after unilateral ligation of the CCA alone or in combination with hypoxia, and partially restored at 24 hours after HI. Moreover, regression analysis showed that the vascularity ratio of ipsilateral hemisphere was moderately correlated with brain infarct size 24 hours after HI, indicating that CBF reduction contributes to of HI brain injury. To further verify the association between CBF and HI-induced brain injury, a neuropeptide C-type natriuretic peptide (CNP) or PBS was intranasally administrated to the brain of mouse pups one hour after HI insult. Brain infarction, CBF imaging and long-term neurobehavioral tests were conducted. The result showed that intranasal administration of CNP preserved ipsilateral CBF, reduced the infarct size, and improved neurological function after HI brain injury. Our findings suggest that CBF alteration is an indicator for neonatal HI brain injury, and 3D ultrasound imaging is a useful non-invasive approach for assessment of HI brain injury in mouse model.
- Published
- 2023
- Full Text
- View/download PDF
10. Mild traumatic brain injury induces microvascular injury and accelerates Alzheimer-like pathogenesis in mice
- Author
-
Yingxi Wu, Haijian Wu, Jianxiong Zeng, Brock Pluimer, Shirley Dong, Xiaochun Xie, Xinying Guo, Tenghuan Ge, Xinyan Liang, Sudi Feng, Youzhen Yan, Jian-Fu Chen, Naomi Sta Maria, Qingyi Ma, Fernando Gomez-Pinilla, and Zhen Zhao
- Subjects
Traumatic brain injury ,Alzheimer’s disease ,Microvascular injury ,Blood–brain barrier ,β-amyloid ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Introduction Traumatic brain injury (TBI) is considered as the most robust environmental risk factor for Alzheimer’s disease (AD). Besides direct neuronal injury and neuroinflammation, vascular impairment is also a hallmark event of the pathological cascade after TBI. However, the vascular connection between TBI and subsequent AD pathogenesis remains underexplored. Methods In a closed-head mild TBI (mTBI) model in mice with controlled cortical impact, we examined the time courses of microvascular injury, blood–brain barrier (BBB) dysfunction, gliosis and motor function impairment in wild type C57BL/6 mice. We also evaluated the BBB integrity, amyloid pathology as well as cognitive functions after mTBI in the 5xFAD mouse model of AD. Results mTBI induced microvascular injury with BBB breakdown, pericyte loss, basement membrane alteration and cerebral blood flow reduction in mice, in which BBB breakdown preceded gliosis. More importantly, mTBI accelerated BBB leakage, amyloid pathology and cognitive impairment in the 5xFAD mice. Discussion Our data demonstrated that microvascular injury plays a key role in the pathogenesis of AD after mTBI. Therefore, restoring vascular functions might be beneficial for patients with mTBI, and potentially reduce the risk of developing AD.
- Published
- 2021
- Full Text
- View/download PDF
11. MicroRNA-210 downregulates TET2 and contributes to inflammatory response in neonatal hypoxic-ischemic brain injury
- Author
-
Qingyi Ma, Chiranjib Dasgupta, Guofang Shen, Yong Li, and Lubo Zhang
- Subjects
Neonatal hypoxia-ischemia ,MicroRNA-210 ,The ten eleven translocation (TET) methylcytosine dioxygenase 2 ,Acetyl-p65 ,Pro-inflammatory cytokines ,BV2 mouse microglia cell line ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Neonatal hypoxic-ischemic (HI) brain injury is a leading cause of acute mortality and chronic disability in newborns. Our previous studies demonstrated that HI insult significantly increased microRNA-210 (miR-210) in the brain of rat pups and inhibition of brain endogenous miR-210 by its inhibitor (LNA) provided neuroprotective effect in HI-induced brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. Methods We made a neonatal HI brain injury model in mouse pups of postnatal day 7 to uncover the mechanism of miR-210 in targeting the ten eleven translocation (TET) methylcytosine dioxygenase 2 that is a transcriptional suppressor of pro-inflammatory cytokine genes in the neonatal brain. TET2 silencing RNA was used to evaluate the role of TET2 in the neonatal HI-induced pro-inflammatory response and brain injury. MiR-210 mimic and inhibitor (LNA) were delivered into the brain of mouse pups to study the regulation of miR-210 on the expression of TET2. Luciferase reporter gene assay was performed to validate the direct binding of miR-210 to the 3′ untranslated region of the TET2 transcript. Furthermore, BV2 mouse microglia cell line was employed to confirm the role of miR-210-TET2 axis in regulating pro-inflammatory response in microglia. Post-assays included chromatin immunoprecipitation (ChIP) assay, co-immunoprecipitation, RT-PCR, brain infarct assay, and neurobehavioral test. Student’s t test or one-way ANOVA was used for statistical analysis. Results HI insult significantly upregulated miR-210, downregulated TET2 protein abundance, and increased NF-κB subunit p65 acetylation level and its DNA binding capacity to the interleukin 1 beta (IL-1β) promoter in the brain of mouse pups. Inhibition of miR-210 rescued TET2 protein level from HI insult and miR-210 mimic decreased TET2 protein level in the brain of mouse pups, suggesting that TET2 is a functional target of miR-210. The co-immunoprecipitation was performed to reveal the role of TET2 in HI-induced inflammatory response in the neonatal brain. The result showed that TET2 interacted with NF-κB subunit p65 and histone deacetylase 3 (HDAC3), a co-repressor of gene transcription. Furthermore, TET2 knockdown increased transcriptional activity of acetyl-p65 on IL-1β gene in the neonatal brain and enhanced HI-induced upregulation of acetyl-p65 level and pro-inflammatory cytokine expression. Of importance, TET2 knockdown exacerbated brain infarct size and neurological deficits and counteracted the neuroprotective effect of miR-210 inhibition. Finally, the in vitro results demonstrated that the miR-210-TET2 axis regulated pro-inflammatory response in BV2 mouse microglia cell line. Conclusions The miR-210-TET2 axis regulates pro-inflammatory cytokine expression in microglia, contributing to neonatal HI brain injury.
- Published
- 2021
- Full Text
- View/download PDF
12. Correction: Blood-brain barrier-associated pericytes internalize and clear aggregated amyloid-β42 by LRP1-dependent apolipoprotein E isoform-specific mechanism
- Author
-
Qingyi Ma, Zhen Zhao, Abhay P. Sagare, Yingxi Wu, Min Wang, Nelly Chuqui Owens, Philip B. Verghese, Joachim Herz, David M. Holtzman, and Berislav V. Zlokovic
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Published
- 2022
- Full Text
- View/download PDF
13. Antenatal Hypoxia Accelerates the Onset of Alzheimer’s Disease Pathology in 5xFAD Mouse Model
- Author
-
Guofang Shen, Shirley Hu, Zhen Zhao, Lubo Zhang, and Qingyi Ma
- Subjects
Alzheimer’s disease ,5xFAD mouse ,antenatal hypoxia ,cognitive impairment ,synapse loss ,gliosis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disorder associated with cognitive impairment and later dementia among the elderly. Mounting evidence shows that adverse maternal environments during the fetal development increase the risk of diseases later in life including neurological disorders, and suggests an early origin in the development of AD-related dementia (ADRD) in utero. In the present study, we investigated the impact of antenatal hypoxia and fetal stress on the initiation of AD-related pathology in offspring of 5xFAD mice. We showed that fetal hypoxia significantly reduced brain and body weight in the fetal and the early postnatal period, which recovered in young adult mice. Using spontaneous Y-maze, novel object recognition (NOR), and open field (OF) tasks, we found that antenatal hypoxia exacerbated cognitive decline in offspring of 5xFAD compared with normoxia control. Of interest, fetal hypoxia did not alter intraneuronal soluble amyloid-β (Aβ) oligomer accumulation in the cortex and hippocampus in 5xFAD mouse offspring, indicating that antenatal hypoxia increased the vulnerability of the brain to synaptotoxic Aβ in the disease onset later in life. Consistent with the early occurrence of cognitive decline, we found synapse loss but not neuronal death in the cerebral cortex in 5xFAD but not wild-type (WT) offspring exposed to antenatal hypoxia. Furthermore, we also demonstrated that antenatal hypoxia significantly increased microglial number and activation, and reactive astrogliosis in the cerebral cortex in WT offspring. Moreover, antenatal hypoxia resulted in an exacerbated increase of microgliosis and astrogliosis in the early stage of AD in 5xFAD offspring. Together, our study reveals a causative link between fetal stress and the accelerated onset of AD-related pathology, and provides mechanistic insights into the developmental origin of aging-related neurodegenerative disorders.
- Published
- 2020
- Full Text
- View/download PDF
14. Blood-brain barrier-associated pericytes internalize and clear aggregated amyloid-β42 by LRP1-dependent apolipoprotein E isoform-specific mechanism
- Author
-
Qingyi Ma, Zhen Zhao, Abhay P Sagare, Yingxi Wu, Min Wang, Nelly Chuqui Owens, Philip B Verghese, Joachim Herz, David M Holtzman, and Berislav V Zlokovic
- Subjects
Pericyte ,Blood-brain barrier (BBB) ,Amyloid-β clearance ,Low-density lipoprotein receptor-related protein 1 (LRP1) ,Apolipoprotein E ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Clearance at the blood-brain barrier (BBB) plays an important role in removal of Alzheimer’s amyloid-β (Aβ) toxin from brain both in humans and animal models. Apolipoprotein E (apoE), the major genetic risk factor for AD, disrupts Aβ clearance at the BBB. The cellular and molecular mechanisms, however, still remain unclear, particularly whether the BBB-associated brain capillary pericytes can contribute to removal of aggregated Aβ from brain capillaries, and whether removal of Aβ aggregates by pericytes requires apoE, and if so, is Aβ clearance on pericytes apoE isoform-specific. Methods We performed immunostaining for Aβ and pericyte biomarkers on brain capillaries (
- Published
- 2018
- Full Text
- View/download PDF
15. Antenatal Hypoxia and Programming of Glucocorticoid Receptor Expression in the Adult Rat Heart
- Author
-
Juanxiu Lv, Qingyi Ma, Chiranjib Dasgupta, Zhice Xu, and Lubo Zhang
- Subjects
hypoxia ,glucocorticoid receptor ,programming ,heart ,DNA methylation ,sex ,Physiology ,QP1-981 - Abstract
Glucocorticoid receptor (GR) signaling is critical for development and function of the heart. Our previous study demonstrated that gestational hypoxia induced epigenetic repression of the GR gene in the developing heart. The present study aims to determine that the alterations of promoter methylation level and epigenetic repression of the GR gene in the developing heart in response to maternal hypoxia is sustained in adult offspring and potential gender differences in the programming of GR gene. Pregnant rats were treated with 10.5% O2 from gestational day 15 (E15) to 21 (E21). Hearts were isolated from 5-month-old male and female offspring with the developing stage being equivalent to 18-year-old human. GR mRNA and protein abundance was determined with real time qRT-PCR and Western blot. GR gene promoter methylation and binding of transcription factors were measured with methylated DNA immunoprecipitation (MeDIP) and Chromatin immunoprecipitation (ChIP). The results showed that antenatal hypoxia significantly decreased the expression of GR mRNA and protein in the hearts of adult male offspring, but not in females, which is ascribed to the differential changes of alternative exon1 mRNA variants of GR gene in male and female hearts in response to prenatal hypoxia. In addition, the downregulation of GR expression in the male heart was correlated with increased methylation levels of CpG dinucleotides in promoters of exon 14, 15, 16, 17, and 110, which resulted in a decrease in the binding of their transcription factors. Thus, the study reveals that antenatal hypoxia results in a reprogramming and long-term change in GR gene expression in the heart by hypermethylation of GR promoter in a sex-differential pattern, which provides a novel mechanism regarding the increased vulnerability of heart later in life with exposure of prenatal hypoxia.
- Published
- 2019
- Full Text
- View/download PDF
16. Inhibition of microRNA-210 provides neuroprotection in hypoxic–ischemic brain injury in neonatal rats
- Author
-
Qingyi Ma, Chiranjib Dasgupta, Yong Li, Nikita M. Bajwa, Fuxia Xiong, Benjamin Harding, Richard Hartman, and Lubo Zhang
- Subjects
Neonatal hypoxic–ischemic brain injury ,MicroRNA-210 ,Glucocorticoid receptor ,Complementary locked nucleic acid (LNA) oligonucleotides ,Intranasal delivery ,Neuroprotection ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Perinatal hypoxic–ischemic encephalopathy (HIE) is associated with high neonatal mortality and severe long-term neurologic morbidity. Yet the mechanisms of brain injury in infants with HIE remain largely elusive. The present study determined a novel mechanism of microRNA-210 (miR-210) in silencing endogenous neuroprotection and increasing hypoxic–ischemic brain injury in neonatal rats. The study further revealed a potential therapeutic effect of miR-210 inhibition using complementary locked nucleic acid oligonucleotides (miR-210-LNA) in 10-day-old neonatal rats in the Rice–Vannucci model. The underlying mechanisms were investigated with intracerebroventricular injection (i.c.v) of miR-210 mimic, miR-210-LNA, glucocorticoid receptor (GR) agonist and antagonist. Luciferase reporter gene assay was conducted for identification of miR-210 targeting GR 3′untranslated region. The results showed that the HI treatment significantly increased miR-210 levels in the brain, and miR-210 mimic significantly decreased GR protein abundance and exacerbated HI brain injury in the pups. MiR-210-LNA administration via i.c.v. 4 h after the HI insult significantly decreased brain miR-210 levels, increased GR protein abundance, reduced HI-induced neuronal death and brain infarct size, and improved long-term neurological function recovery. Of importance, the intranasal delivery of miR-210-LNA 4 h after the HI insult produced similar effects in decreasing HI-induced neonatal brain injury and improving neurological function later in life. Altogether, the present study provides evidence of a novel mechanism of miR-210 in a neonatal HI brain injury model, and suggests a potential therapeutic approach of miR-210 inhibition in the treatment of neonatal HIE.
- Published
- 2016
- Full Text
- View/download PDF
17. RETRACTED ARTICLE: Pericyte loss influences Alzheimer-like neurodegeneration in mice
- Author
-
Abhay P. Sagare, Robert D. Bell, Zhen Zhao, Qingyi Ma, Ethan A. Winkler, Anita Ramanathan, and Berislav V. Zlokovic
- Subjects
Science - Abstract
Abstract Pericytes are cells in the blood–brain barrier that degenerate in Alzheimer’s disease (AD), a neurological disorder associated with neurovascular dysfunction, abnormal elevation of amyloid β-peptide (Aβ), tau pathology and neuronal loss. Whether pericyte degeneration can influence AD-like neurodegeneration and contribute to disease pathogenesis remains, however, unknown. Here we show that in mice overexpressing Aβ-precursor protein, pericyte loss elevates brain Aβ40 and Aβ42 levels and accelerates amyloid angiopathy and cerebral β-amyloidosis by diminishing clearance of soluble Aβ40 and Aβ42 from brain interstitial fluid prior to Aβ deposition. We further show that pericyte deficiency leads to the development of tau pathology and an early neuronal loss that is normally absent in Aβ-precursor protein transgenic mice, resulting in cognitive decline. Our data suggest that pericytes control multiple steps of AD-like neurodegeneration pathogenic cascade in Aβ-precursor protein-overexpressing mice. Therefore, pericytes may represent a novel therapeutic target to modify disease progression in AD.
- Published
- 2013
- Full Text
- View/download PDF
18. P2X7R/cryopyrin inflammasome axis inhibition reduces neuroinflammation after SAH
- Author
-
Sheng Chen, Qingyi Ma, Paul R. Krafft, Qin Hu, William Rolland, II, Prativa Sherchan, Jianmin Zhang, Jiping Tang, and John H. Zhang
- Subjects
Subarachnoid hemorrhage ,Early brain injury ,Cryopyrin ,P2X purinoceptor 7 ,Inflammation ,Edema ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Neuroinflammation contributes to the pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Cytotoxic events following SAH, such as extracellular accumulation of adenosine triphosphate (ATP), may activate the P2X purinoceptor 7 (P2X7R)/cryopyrin inflammasome axis, thus inducing the proinflammatory cytokine IL-1β/IL-18 secretion. We therefore hypothesized that inhibition of P2X7R/cryopyrin inflammasome axis would ameliorate neuroinflammation after SAH. In the present study, SAH was induced by the endovascular perforation in rats. Small interfering RNAs (siRNAs) of P2X7R or cryopyrin were administered intracerebroventricularly 24 h before SAH. Brilliant blue G (BBG), a non-competitive antagonist of P2X7R, was administered intraperitoneally 30 min following SAH. Post-assessments including SAH severity score, neurobehavioral test, brain water content, Western blot and immunofluorescence, were performed. Administration of P2X7R and cryopyrin siRNA as well as pharmacologic blockade of P2X7R by BBG ameliorated neurological deficits and brain edema at 24 h following SAH. Inhibition of P2X7R/cryopyrin inflammasome axis suppressed caspase-1 activation, which subsequently decreased maturation of IL-1β/IL-18. To investigate the link between P2X7R and cryopyrin inflammasome in vivo, Benzoylbenzoyl-ATP (BzATP), a P2X7R agonist, was given to lipopolysaccharide (LPS) primed naive rats with scramble or cryopyrin siRNAs. In LPS-primed naive rats, BzATP induced caspase-1 activation and mature IL-1β release were neutralized by cryopyrin siRNA. Thus, the P2X7R/cryopyrin inflammasome axis may contribute to neuroinflammation via activation of caspase-1 and thereafter mature IL-1β/IL-18 production following SAH. Therapeutic interventions targeting P2X7R/cryopyrin pathway may be a novel approach to ameliorate EBI following SAH.
- Published
- 2013
- Full Text
- View/download PDF
19. PAR-1 antagonist SCH79797 ameliorates apoptosis following surgical brain injury through inhibition of ASK1-JNK in rats
- Author
-
Anatol Manaenko, Xuejun Sun, Cherine H. Kim, Junhao Yan, Qingyi Ma, and John H. Zhang
- Subjects
Surgical brain injury ,Thrombin ,PAR-1 ,Apoptosis ,ASK1 ,JNK ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Neurosurgical procedures inevitably produce intraoperative hemorrhage. The subsequent entry of blood into the brain parenchyma results in the release of large amounts of thrombin, a known contributor to perihematomal edema formation and apoptosis after brain injury. The present study seeks to test 1) the effect of surgically induced brain injury (SBI) on thrombin activity, expression of thrombin's receptor PAR-1, and PAR-1 mediated apoptosis; 2) the effect of thrombin inhibition by argatroban and PAR-1 inhibition by SCH79797 on the development of secondary brain injury in the SBI model on rats.A total of 88 Sprague–Dawley male rats were randomly divided into sham, vehicle-, argatroban-, or SCH79797-treated groups. SBI involved partial resection of the right frontal lobe under inhalation isoflurane anesthesia. Sham-operated animals received only craniotomy. Thrombin activity, brain water content, and neurological deficits were measured at 24 h following SBI. Involvement of the Ask1/JNK pathway in PAR-1-induced post-SBI apoptosis was characterized by using Ask1 or JNK inhibitors.We observed that SBI increased thrombin activity, yet failed to demonstrate any effect on PAR-1 expression. Argatroban and SCH79797 reduced SBI-induced brain edema and neurological deficits in a dose-dependent manner. SBI-induced apoptosis seemed mediated by the PAR-1/Ask1/JNK pathways. Administration of SCH79797 ameliorated the apoptosis following SBI.Our findings indicate that PAR-1 antagonist protects against secondary brain injury after SBI by decreasing both brain edema and apoptosis by inactivating PAR-1/Ask1/JNK pathway. The anti-apoptotic effect of PAR-1 antagonists may provide a promising path for therapy following SBI.
- Published
- 2013
- Full Text
- View/download PDF
20. Fibroblast growth factors preserve blood–brain barrier integrity through RhoA inhibition after intracerebral hemorrhage in mice
- Author
-
Bin Huang, Paul R. Krafft, Qingyi Ma, William B. Rolland, Basak Caner, Tim Lekic, Anatol Manaenko, Mai Le, Jiping Tang, and John H. Zhang
- Subjects
Fibroblast growth factor ,Thrombin ,Rac1 ,RhoA ,Intracerebral hemorrhage ,Brain injury ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Fibroblast growth factors (FGFs) maintain and promote vascular integrity; however whether FGFs protect the blood–brain barrier (BBB) after intracerebral hemorrhage (ICH) remains unexplored. In this present study, we hypothesized that exogenous FGF administration attenuates brain injury after ICH, specifically by preserving endothelial adherens junctions, therefore reducing vasogenic brain edema and attenuating neurofunctional deficits in mice subjected to experimental ICH.Acid fibroblast growth factor (FGF1) or basic fibroblast growth factor (FGF2) was administered intracerebroventricularly (ICV) at 0.5 h after intrastriatal injection of bacterial collagenase (cICH) or autologous whole blood (bICH). Fibroblast growth factor receptor (FGFR) inhibitor PD173074 and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 were additionally administered with FGF2. The selective Rho-associated coiled-coil forming protein serine/threonine kinase (ROCK) inhibitor Y27632 was independently administered at 0.5 h after cICH. Brain water content and neurofunctional deficits were evaluated at 24 and 72 h after ICH induction. Evans blue extravasation as well as Western blot analysis for the quantification of activated FGFR, Akt, Ras-related C3 botulinum toxin substrate 1 (Rac1), Ras homolog gene family member A (RhoA) and adherens junction proteins (p120-catenin, β-catenin and VE-cadherin) were conducted at 72 h post-cICH. FGF treatment reduced perihematomal brain edema and improved neurofunctional deficits at 72 h after experimental ICH (p
- Published
- 2012
- Full Text
- View/download PDF
21. Atp13a5 Marker Reveals Pericyte Specification in the Mouse Central Nervous System.
- Author
-
Xinying Guo, Shangzhou Xia, Tenghuan Ge, Yangtao Lin, Shirley Hu, Haijian Wu, Xiaochun Xie, Bangyan Zhang, Zhang, Sonia, Jianxiong Zeng, Jian-Fu Chen, Montagne, Axel, Fan Gao, Qingyi Ma, and Zhen Zhao
- Subjects
CARDIOVASCULAR system ,PERICYTES ,CENTRAL nervous system ,BLOOD-brain barrier ,GENETIC models ,SPINAL cord - Abstract
Perivascular mural cells including vascular smooth cells (VSMCs) and pericytes are integral components of the vascular system. In the central nervous system (CNS), pericytes are also indispensable for the blood-brain barrier (BBB), blood-spinal cord barrier, and blood-retinal barrier and play key roles in maintaining cerebrovascular and neuronal functions. However, the functional specifications of pericytes between CNS and peripheral organs have not been resolved at the genetic and molecular levels. Hence, the generation of reliable CNS pericyte-specific models and genetic tools remains very challenging. Here, we report a new CNS pericyte marker in mice. This putative cation-transporting ATPase 13A5 (Atp13a5)marker was identified through single-cell transcriptomics, based on its specificity to brain pericytes. We further generated a knock-inmodel with both tdTomato reporter and Cre recombinase. Using this model to trace the distribution of Atp13a5-positive pericytes in mice, we found that the tdTomato reporter reliably labels the CNS pericytes, including the ones in spinal cord and retina but not peripheral organs. Interestingly, brain pericytes are likely shaped by the developing neural environment, as Atp13a5-positive pericytes start to appear around murine embryonic day 15 (E15) and expand along the cerebrovasculature. Thus, Atp13a5 is a specific marker of CNS pericyte lineage, and this Atp13a5-based model is a reliable tool to explore the heterogeneity of pericytes and BBB functions in health and diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. MicroRNA-210 Downregulates TET2 (Ten-Eleven Translocation Methylcytosine Dioxygenase 2) and Contributes to Neuroinflammation in Ischemic Stroke of Adult Mice
- Author
-
Yong Li, Rui Song, Guofang Shen, Lei Huang, DaLiao Xiao, Qingyi Ma, and Lubo Zhang
- Subjects
Advanced and Specialized Nursing ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine - Abstract
Background: Stroke is a leading cause of morbidity and mortality worldwide. Neuroinflammation plays a key role in acute brain injury of ischemic stroke. MicroRNA-210 (miR210) is the master hypoxamir and regulates microglial activation and inflammation in a variety of diseases. In this study, we uncovered the mechanism of miR210 in orchestrating ischemic stroke–induced neuroinflammation through repression of TET2 (ten-eleven translocation methylcytosine dioxygenase 2) in the adult mouse brain. Methods: Ischemic stroke was induced in adult WT (wild type) or miR210 KO (miR210 deficient) mice by transient intraluminal middle cerebral artery occlusion. Injection of TET2 silencing RNA or miR210 complementary locked nucleic acid oligonucleotides, or miR210 KO mice were used to validate miR210-TET2 axis and its role in ischemic brain injury. Furthermore, the effect of TET2 overexpression on miR210-stimulated proinflammatory cytokines was examined in BV2 microglia. Post assays included magnetic resonance imaging scan for brain infarct size; neurobehavioral tests, reverse transcription-quantitative polymerase chain reaction, and Western blot for miR210; and TET2 levels, flow cytometry, and ELISA for neuroinflammation in the brain after stroke or microglia in vitro. Results: miR210 injection significantly reduced TET2 protein abundance in the brain, while miR210 complementary locked nucleic acid oligonucleotides or miR210 KO preserved TET2 regardless of ischemic brain injury. TET2 knockdown reversed the protective effects of miR210 inhibition or miR210 KO on ischemic stroke–induced brain infarct size and neurobehavioral deficits. Moreover, flow cytometry and ELISA assays showed that TET2 knockdown also significantly dampened the anti-inflammatory effect of miR210 inhibition on microglial activation and IL (interleukin)-6 release after stroke. In addition, overexpression of TET2 in BV2 microglia counteracted miR210-induced increase in cytokines. Conclusions: miR210 inhibition reduced ischemic stroke–induced neuroinflammatory response via repression of TET2 in the adult mouse brain, suggesting that miR210 is a potential treatment target for acute brain injury after ischemic stroke.
- Published
- 2023
- Full Text
- View/download PDF
23. Multi-omics analyzes of Rosa gigantea illuminate tea scent biosynthesis and release mechanisms
- Author
-
Lijun Zhou, Sihui Wu, Yunyi Chen, Runhuan Huang, Bixuan Cheng, Qingyi Mao, Tinghan Liu, Yuchen Liu, Kai Zhao, Huitang Pan, Chao Yu, Xiang Gao, Le Luo, and Qixiang Zhang
- Subjects
Science - Abstract
Abstract Rose is an important ornamental crop cultivated globally for perfume production. However, our understanding of the mechanisms underlying scent production and molecular breeding for fragrance is hindered by the lack of a reference genome for tea roses. We present the first complete telomere-to-telomere (T2T) genome of Rosa gigantea, with high quality (QV > 60), including detailed characterization of the structural features of repetitive regions. The expansion of genes associated with phenylpropanoid biosynthesis may account for the unique tea scent. We uncover the release rhythm of aromatic volatile organic compounds and their gene regulatory networks through comparative genomics and time-ordered gene co-expression networks. Analyzes of eugenol homologs demonstrate how plants attract pollinators using specialized phenylpropanoids in specific tissues. This study highlights the conservation and utilization of genetic diversity from wild endangered species through multi-omics approaches, providing a scientific foundation for enhancing rose fragrance via de novo domestication.
- Published
- 2024
- Full Text
- View/download PDF
24. MicroRNAs in the Blood-Brain Barrier in Hypoxic-Ischemic Brain Injury
- Author
-
Qingyi Ma and Guofang Shen
- Subjects
0301 basic medicine ,tight junction ,Hypoxic ischemic brain injury ,Blood–brain barrier ,Article ,Brain ischemia ,03 medical and health sciences ,0302 clinical medicine ,microRNA ,medicine ,ischemic stroke ,Humans ,Pharmacology (medical) ,BBB disruption ,Pathological ,miRNA ,Pharmacology ,Tight junction ,MMP ,business.industry ,Infant, Newborn ,Biological Transport ,General Medicine ,Hypoxia (medical) ,Hypoxic ischemia encephalopathy (HIE) ,medicine.disease ,neural inflammation ,Psychiatry and Mental health ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Blood-Brain Barrier ,Brain Injuries ,Hypoxia-Ischemia, Brain ,Chronic disability ,Neurology (clinical) ,medicine.symptom ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Hypoxic-ischemic (HI) brain injury is a leading cause of acute mortality and chronic disability in newborns. Current evidence shows that cerebral microvascular response and compromised blood-brain barrier (BBB) integrity occur rapidly and could primarily be responsible for the brain injury observed in many infants with HI brain injury. MicroRNAs (miRNAs) are a type of highly conserved non-coding RNAs (ncRNAs), which consist of 21-25 nucleotides in length and usually lead to suppression of target gene expression. Growing evidence has revealed that brainenriched miRNAs act as versatile regulators of BBB dysfunctions in various neurological disorders including neonatal HI brain injury. In the present review, we summarize the current findings regarding the role of miRNAs in BBB impairment after hypoxia/ischemia brain injury. Specifically, we focus on the recent progress of miRNAs in the pathologies of neonatal HI brain injury. These findings can not only deepen our understanding of the role of miRNAs in BBB impairment in HI brain injury, but also provide insight into the development of new therapeutic strategies for preservation of BBB integrity under pathological conditions.
- Published
- 2020
25. MicroRNAs in brain development and cerebrovascular pathophysiology
- Author
-
Lubo Zhang, William J. Pearce, and Qingyi Ma
- Subjects
0301 basic medicine ,Brain development ,Physiology ,Neurogenesis ,Neovascularization, Physiologic ,Hypoxic ischemic brain injury ,Biology ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,microRNA ,Animals ,Humans ,Neurons ,Neovascularization, Pathologic ,Brain ,Gene Expression Regulation, Developmental ,Cell Biology ,Pathophysiology ,Cerebrovascular Disorders ,MicroRNAs ,030104 developmental biology ,Hypoxia-Ischemia, Brain ,Theme ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
MicroRNAs (miRNAs) are a class of highly conserved non-coding RNAs with 21–25 nucleotides in length and play an important role in regulating gene expression at the posttranscriptional level via base-paring with complementary sequences of the 3′-untranslated region of the target gene mRNA, leading to either transcript degradation or translation inhibition. Brain-enriched miRNAs act as versatile regulators of brain development and function, including neural lineage and subtype determination, neurogenesis, synapse formation and plasticity, neural stem cell proliferation and differentiation, and responses to insults. Herein, we summarize the current knowledge regarding the role of miRNAs in brain development and cerebrovascular pathophysiology. We review recent progress of the miRNA-based mechanisms in neuronal and cerebrovascular development as well as their role in hypoxic-ischemic brain injury. These findings hold great promise, not just for deeper understanding of basic brain biology but also for building new therapeutic strategies for prevention and treatment of pathologies such as cerebral ischemia.
- Published
- 2019
- Full Text
- View/download PDF
26. MicroRNA-210 Downregulates ISCU and Induces Mitochondrial Dysfunction and Neuronal Death in Neonatal Hypoxic-Ischemic Brain Injury
- Author
-
Lubo Zhang, Chiranjib Dasgupta, Lei Huang, Yong Li, and Qingyi Ma
- Subjects
Iron-Sulfur Proteins ,0301 basic medicine ,Cell Survival ,Neuroscience (miscellaneous) ,Down-Regulation ,Oxidative phosphorylation ,medicine.disease_cause ,Neuroprotection ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Animals ,Gene silencing ,Cells, Cultured ,Neurons ,chemistry.chemical_classification ,Reactive oxygen species ,Gene knockdown ,Cell Death ,biology ,Mitochondria ,Rats ,Up-Regulation ,Cell biology ,Oxygen ,MicroRNAs ,Oxidative Stress ,Glucose ,030104 developmental biology ,Animals, Newborn ,Neurology ,chemistry ,Brain Injuries ,Hypoxia-Ischemia, Brain ,biology.protein ,ISCU ,Reactive Oxygen Species ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Neonatal hypoxic-ischemic (HI) brain injury causes significant mortality and long-term neurologic sequelae. We previously demonstrated that HI significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of brain endogenous miR-210 was neuroprotective in HI brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. Using both in vivo and in vitro models, herein we uncover a novel mechanism mediating oxidative brain injury after neonatal HI, in which miR-210 induces mitochondrial dysfunction via downregulation of iron-sulfur cluster assembly protein (ISCU). Inhibition of miR-210 significantly ameliorates mitochondrial dysfunction, oxidative stress, and neuronal loss in the neonatal brain subjected to HI, as well as in primary cortical neurons exposed to oxygen-glucose deprivation (OGD). These effects are mediated through ISCU, in that miR-210 mimic decreases ISCU abundance in the brains of rat pups and primary cortical neurons, and inhibition of miR-210 protects ISCU against HI in vivo or OGD in vitro. Deletion of miR-210 binding sequences at the 3′UTR of ISCU transcript ablates miR-210-induced downregulation of ISCU protein abundance in PC12 cells. In primary cortical neurons, miR-210 mimic or silencing ISCU results in mitochondrial dysfunction, reactive oxygen species production, and activation of caspase-dependent death pathways. Of importance, knockdown of ISCU increases HI-induced injury in the neonatal rat brain and counteracts the neuroprotection of miR-210 inhibition. Therefore, miR-210 by downregulating ISCU and inducing mitochondrial dysfunction in neurons is a potent contributor of oxidative brain injury after neonatal HI.
- Published
- 2019
- Full Text
- View/download PDF
27. Mild traumatic brain injury induces microvascular injury and accelerates Alzheimer-like pathogenesis in mice
- Author
-
Shirley Dong, Tenghuan Ge, Yingxi Wu, Xiaochun Xie, Xinying Guo, Qingyi Ma, Jian-Fu Chen, Haijian Wu, Sudi Feng, Naomi S Sta Maria, Jianxiong Zeng, Brock Pluimer, Youzhen Yan, Fernando Gomez-Pinilla, Zhen Zhao, and Xinyan Liang
- Subjects
Pathology ,Aging ,Neurology ,Neurodegenerative ,Inbred C57BL ,Alzheimer's Disease ,Transgenic ,Pathogenesis ,Mice ,Traumatic brain injury ,Injury - Trauma - (Head and Spine) ,Medicine ,2.1 Biological and endogenous factors ,Aetiology ,β ,β-amyloid ,medicine.anatomical_structure ,Cerebral blood flow ,Neurological ,Disease Progression ,amyloid ,Pericyte ,medicine.symptom ,Alzheimer’s disease ,medicine.medical_specialty ,Microvascular injury ,Clinical Sciences ,Mice, Transgenic ,Blood–brain barrier ,Blood– ,Pathology and Forensic Medicine ,s disease ,Cellular and Molecular Neuroscience ,Alzheimer Disease ,Acquired Cognitive Impairment ,Animals ,Humans ,Cognitive Dysfunction ,RC346-429 ,Alzheimer’ ,Neuroinflammation ,Brain Concussion ,brain barrier ,business.industry ,Research ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,medicine.disease ,Brain Disorders ,Mice, Inbred C57BL ,nervous system ,Gliosis ,Microvessels ,Injury (total) Accidents/Adverse Effects ,Dementia ,Neurology. Diseases of the nervous system ,Neurology (clinical) ,Biochemistry and Cell Biology ,business ,Injury - Traumatic brain injury - Abstract
Introduction Traumatic brain injury (TBI) is considered as the most robust environmental risk factor for Alzheimer’s disease (AD). Besides direct neuronal injury and neuroinflammation, vascular impairment is also a hallmark event of the pathological cascade after TBI. However, the vascular connection between TBI and subsequent AD pathogenesis remains underexplored. Methods In a closed-head mild TBI (mTBI) model in mice with controlled cortical impact, we examined the time courses of microvascular injury, blood–brain barrier (BBB) dysfunction, gliosis and motor function impairment in wild type C57BL/6 mice. We also evaluated the BBB integrity, amyloid pathology as well as cognitive functions after mTBI in the 5xFAD mouse model of AD. Results mTBI induced microvascular injury with BBB breakdown, pericyte loss, basement membrane alteration and cerebral blood flow reduction in mice, in which BBB breakdown preceded gliosis. More importantly, mTBI accelerated BBB leakage, amyloid pathology and cognitive impairment in the 5xFAD mice. Discussion Our data demonstrated that microvascular injury plays a key role in the pathogenesis of AD after mTBI. Therefore, restoring vascular functions might be beneficial for patients with mTBI, and potentially reduce the risk of developing AD.
- Published
- 2021
28. Endothelial LRP1 protects against neurodegeneration by blocking cyclophilin A
- Author
-
Kassandra Kisler, Zhonghua Dai, Mikko T Huuskonen, Qingyi Ma, Zhen Zhao, Yaoming Wang, Sanket V Rege, Joachim Herz, Abhay P. Sagare, Jakob Körbelin, Berislav V. Zlokovic, Axel Montagne, and Angeliki M. Nikolakopoulou
- Subjects
Male ,0301 basic medicine ,Cell signaling ,Endothelium ,Angiogenesis ,Immunology ,Mice, Transgenic ,Article ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,Cyclophilin A ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Animals ,Immunology and Allergy ,Cognitive Dysfunction ,Enzyme Inhibitors ,Cells, Cultured ,Cyclophilin ,Neurons ,Tight junction ,Chemistry ,Neurodegeneration ,Endothelial Cells ,Genetic Therapy ,medicine.disease ,LRP1 ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Matrix Metalloproteinase 9 ,Blood-Brain Barrier ,Cyclosporine ,cardiovascular system ,Female ,Low Density Lipoprotein Receptor-Related Protein-1 ,030217 neurology & neurosurgery ,Signal Transduction ,Neuroscience - Abstract
Endothelial low-density lipoprotein receptor–related protein 1 (LRP1) protects against neurodegeneration by inhibiting the proinflammatory cyclophilin A–matrix metalloproteinase-9 pathway at the blood–brain barrier. These findings have implications for the pathophysiology and treatment of neurodegenerative disorders linked to vascular dysfunction., The low-density lipoprotein receptor–related protein 1 (LRP1) is an endocytic and cell signaling transmembrane protein. Endothelial LRP1 clears proteinaceous toxins at the blood–brain barrier (BBB), regulates angiogenesis, and is increasingly reduced in Alzheimer’s disease associated with BBB breakdown and neurodegeneration. Whether loss of endothelial LRP1 plays a direct causative role in BBB breakdown and neurodegenerative changes remains elusive. Here, we show that LRP1 inactivation from the mouse endothelium results in progressive BBB breakdown, followed by neuron loss and cognitive deficits, which is reversible by endothelial-specific LRP1 gene therapy. LRP1 endothelial knockout led to a self-autonomous activation of the cyclophilin A–matrix metalloproteinase-9 pathway in the endothelium, causing loss of tight junctions underlying structural BBB impairment. Cyclophilin A inhibition in mice with endothelial-specific LRP1 knockout restored BBB integrity and reversed and prevented neuronal loss and behavioral deficits. Thus, endothelial LRP1 protects against neurodegeneration by inhibiting cyclophilin A, which has implications for the pathophysiology and treatment of neurodegeneration linked to vascular dysfunction.
- Published
- 2021
- Full Text
- View/download PDF
29. MicroRNA-210 downregulates TET2 and contributes to inflammatory response in neonatal hypoxic-ischemic brain injury
- Author
-
Lubo Zhang, Guofang Shen, Yong Li, Qingyi Ma, and Chiranjib Dasgupta
- Subjects
Male ,Pro-inflammatory cytokines ,Immunology ,Down-Regulation ,Biology ,Neuroprotection ,lcsh:RC346-429 ,Cell Line ,Dioxygenases ,Proinflammatory cytokine ,The ten eleven translocation (TET) methylcytosine dioxygenase 2 ,Mice ,Cellular and Molecular Neuroscience ,Neonatal hypoxia-ischemia ,Downregulation and upregulation ,Proto-Oncogene Proteins ,microRNA ,medicine ,Animals ,lcsh:Neurology. Diseases of the nervous system ,Gene knockdown ,Microglia ,Research ,General Neuroscience ,Acetyl-p65 ,HDAC3 ,Cell biology ,DNA-Binding Proteins ,MicroRNAs ,Liposaccharide (LPS) ,medicine.anatomical_structure ,Animals, Newborn ,Neurology ,Hypoxia-Ischemia, Brain ,Female ,Inflammation Mediators ,MicroRNA-210 ,Chromatin immunoprecipitation ,BV2 mouse microglia cell line - Abstract
Background Neonatal hypoxic-ischemic (HI) brain injury is a leading cause of acute mortality and chronic disability in newborns. Our previous studies demonstrated that HI insult significantly increased microRNA-210 (miR-210) in the brain of rat pups and inhibition of brain endogenous miR-210 by its inhibitor (LNA) provided neuroprotective effect in HI-induced brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. Methods We made a neonatal HI brain injury model in mouse pups of postnatal day 7 to uncover the mechanism of miR-210 in targeting the ten eleven translocation (TET) methylcytosine dioxygenase 2 that is a transcriptional suppressor of pro-inflammatory cytokine genes in the neonatal brain. TET2 silencing RNA was used to evaluate the role of TET2 in the neonatal HI-induced pro-inflammatory response and brain injury. MiR-210 mimic and inhibitor (LNA) were delivered into the brain of mouse pups to study the regulation of miR-210 on the expression of TET2. Luciferase reporter gene assay was performed to validate the direct binding of miR-210 to the 3′ untranslated region of the TET2 transcript. Furthermore, BV2 mouse microglia cell line was employed to confirm the role of miR-210-TET2 axis in regulating pro-inflammatory response in microglia. Post-assays included chromatin immunoprecipitation (ChIP) assay, co-immunoprecipitation, RT-PCR, brain infarct assay, and neurobehavioral test. Student’s t test or one-way ANOVA was used for statistical analysis. Results HI insult significantly upregulated miR-210, downregulated TET2 protein abundance, and increased NF-κB subunit p65 acetylation level and its DNA binding capacity to the interleukin 1 beta (IL-1β) promoter in the brain of mouse pups. Inhibition of miR-210 rescued TET2 protein level from HI insult and miR-210 mimic decreased TET2 protein level in the brain of mouse pups, suggesting that TET2 is a functional target of miR-210. The co-immunoprecipitation was performed to reveal the role of TET2 in HI-induced inflammatory response in the neonatal brain. The result showed that TET2 interacted with NF-κB subunit p65 and histone deacetylase 3 (HDAC3), a co-repressor of gene transcription. Furthermore, TET2 knockdown increased transcriptional activity of acetyl-p65 on IL-1β gene in the neonatal brain and enhanced HI-induced upregulation of acetyl-p65 level and pro-inflammatory cytokine expression. Of importance, TET2 knockdown exacerbated brain infarct size and neurological deficits and counteracted the neuroprotective effect of miR-210 inhibition. Finally, the in vitro results demonstrated that the miR-210-TET2 axis regulated pro-inflammatory response in BV2 mouse microglia cell line. Conclusions The miR-210-TET2 axis regulates pro-inflammatory cytokine expression in microglia, contributing to neonatal HI brain injury.
- Published
- 2021
- Full Text
- View/download PDF
30. Additional file 1 of Mild traumatic brain injury induces microvascular injury and accelerates Alzheimer-like pathogenesis in mice
- Author
-
Yingxi Wu, Haijian Wu, Jianxiong Zeng, Pluimer, Brock, Dong, Shirley, Xiaochun Xie, Xinying Guo, Tenghuan Ge, Xinyan Liang, Sudi Feng, Youzhen Yan, Chen, Jian-Fu, Maria, Naomi Sta, Qingyi Ma, Gomez-Pinilla, Fernando, and Zhao, Zhen
- Abstract
Additional file 1: Supplementary figures. Supplementary Fig. 1 Traumatic brain injury models with controlled cortical impact in mice. (A) The timeline for the establishment of the TBI mouse model and the other experimental procedures. (B) Brain precision impactor device (RWD Life Science) to establish mTBI mouse model. (C-D) The experimental parameters used in the establishment of TBI mouse models. Supplementary Fig. 2 Neuropathological changes 3 days after sTBI or mTBI in mouse model. (A) Cerebral edema measured by brain water content. The injury significantly induced the development of cerebral edema at 24 hours post-injury in sTBI model compared to mTBI model. n = 8 mice per group, Data are presented as Mean ± SD; ***, P < 0.001; NS, non-significant (P > 0.05), one-way ANOVA followed by Bonferroni′s post-hoc tests. (B) Representative confocal images showing NeuN (green) staining and TUNEL assay (red) for apoptotic neuronal death in the impacted ipsilateral area after sTBI and mTBI, or sham-operation. Scale bar, 50 μm. (C) Quantification of the percentage of TUNEL-positive neuronal death in sTBI (n = 6 mice) or mTBI (n = 6 mice). Data are presented as Mean ± SD; *** P < 0.001 by Student′s t-test. Dash line indicates an average value from sham-operated group (n = 3 mice). (D-E) Representative immunoblots (D) and quantification (E) of Synapsin I from the cortex in the injury ipsilateral side of sham-operated, mTBI and sTBI mice. β-Actin: loading control. Data are presented as Mean ± SD; n = 3 mice per group respectively; ***, P < 0.001; **, P < 0.01; one-way ANOVA followed by Bonferroni′s post-hoc tests. Supplementary Fig. 3 Neuroinflammatory changes 3 days after sTBI or mTBI in mouse model. (A) Representative images of fluorescence immunostaining for Iba1 (gray) and DAPI (blue), boxed regions show an activated and phagocytotic microglia (a1, a3) in the impacted ipsilateral side, and ramified microglia at resting state (a2, a4) in the contralateral side. Scale bar in (A) 50 μm, in (a1-a6) 10 μm. (B) Quantification for the percentage of activated Iba1 positive cells (n = 6 mice per group) in the impact side (ipsi) and contralateral side (contra) in sTBI and mTBI mice (see method). Dash line indicates the average value from the sham-operated group (n = 5 mice). (C) Representative images of fluorescence immunostaining for GFAP (green) and DAPI (blue), boxed regions (c1-c4) show astrocytes in the impact side and the contralateral side. Scale bar in (C) 50 μm, in (c1-c6) 10 μm. (D) Quantification for the GFAP positive cells per mm2 (n = 6 mice per group) in the impact side (ipsi) and contralateral side (contra) in sTBI and mTBI mice. Dash line indicates the average value from sham-operated group (n = 5 mice). Data are presented as Mean ± SD; ***, P < 0.001; **, P < 0.01; *, P < 0.05; NS, non-significant (P > 0.05) by by Student's t-test. Supplementary Fig. 4 Vascular impairment in the subacute phase of mTBI in mice. (A-B) Representative images (A) and quantification (B) showing extravascular fibrin deposits in brain parenchyma 3 days after mTBI. Lectin (gray), fibrin (red). n = 7 mice; Scale bar = 20 μm. (C-D) Representative images (C) and quantification (D) showing extravasation of intravenously administrated Alexa-555 cadaverine (red) in brain parenchyma 3 days after mTBI. Lectin (green), n = 7 mice, scale bar = 50 μm. In B, D, data are presented as Mean ± SD; ***, P < 0.001 by Student's t-test. Dash lines indicate average value from sham-operated group (n = 3 mice). (E-F) Cerebral blood flow reduction 3 days after mTBI. Representative laser speckle contrast imaging (LSCI) images (E) and quantification of boxed regions (F) showing blood flow changes in the cortical regions 3 days after mTBI. Scale bar = 50 μm. Supplementary Fig. 5 The time course of microglial activation after mTBI in mice. (A) Representative confocal microscope images showing Iba1-positive microglia cells (red) in cortex (CTX), Cornu Ammonis 1 (CA1) and dentate gyrus (DG) area 1-day, 3- and 8- days post operation (DPO). Scale bar = 100 μm. (B-D) Quantification for the Iba1-positive microglia cells per mm2 (n = 5 mice per time point) in CTX, CA1 and DG area 1 day, 3- and 8-days post operation (DPO). Data are presented as Mean ± SD; ***, P < 0.001; **, P < 0.01; one-way ANOVA followed by Bonferroni's post-hoc tests.
- Published
- 2021
- Full Text
- View/download PDF
31. Antenatal Hypoxia Accelerates the Onset of Alzheimer’s Disease Pathology in 5xFAD Mouse Model
- Author
-
Shirley Hu, Zhen Zhao, Qingyi Ma, Lubo Zhang, and Guofang Shen
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Aging ,Offspring ,Cognitive Neuroscience ,lcsh:RC321-571 ,03 medical and health sciences ,0302 clinical medicine ,5xFAD mouse ,medicine ,synapse loss ,Dementia ,Cognitive decline ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Original Research ,cognitive impairment ,Fetus ,business.industry ,Hypoxia (medical) ,medicine.disease ,Astrogliosis ,030104 developmental biology ,medicine.anatomical_structure ,gliosis ,Gliosis ,Cerebral cortex ,antenatal hypoxia ,medicine.symptom ,business ,Alzheimer’s disease ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder associated with cognitive impairment and later dementia among the elderly. Mounting evidence shows that adverse maternal environments during the fetal development increase the risk of diseases later in life including neurological disorders, and suggests an early origin in the development of AD-related dementia (ADRD) in utero. In the present study, we investigated the impact of antenatal hypoxia and fetal stress on the initiation of AD-related pathology in offspring of 5xFAD mice. We showed that fetal hypoxia significantly reduced brain and body weight in the fetal and the early postnatal period, which recovered in young adult mice. Using spontaneous Y-maze, novel object recognition (NOR), and open field (OF) tasks, we found that antenatal hypoxia exacerbated cognitive decline in offspring of 5xFAD compared with normoxia control. Of interest, fetal hypoxia did not alter intraneuronal soluble amyloid-β (Aβ) oligomer accumulation in the cortex and hippocampus in 5xFAD mouse offspring, indicating that antenatal hypoxia increased the vulnerability of the brain to synaptotoxic Aβ in the disease onset later in life. Consistent with the early occurrence of cognitive decline, we found synapse loss but not neuronal death in the cerebral cortex in 5xFAD but not wild-type (WT) offspring exposed to antenatal hypoxia. Furthermore, we also demonstrated that antenatal hypoxia significantly increased microglial number and activation, and reactive astrogliosis in the cerebral cortex in WT offspring. Moreover, antenatal hypoxia resulted in an exacerbated increase of microgliosis and astrogliosis in the early stage of AD in 5xFAD offspring. Together, our study reveals a causative link between fetal stress and the accelerated onset of AD-related pathology, and provides mechanistic insights into the developmental origin of aging-related neurodegenerative disorders.
- Published
- 2020
- Full Text
- View/download PDF
32. Microvascular Injury in Mild Traumatic Brain Injury Accelerates Alzheimer-like Pathogenesis in Mice
- Author
-
Haijian Wu, Jianxiong Zeng, Brock Pluimer, Yingxi Wu, Xinying Guo, Youzhen Yan, Fernando Gomez-Pinilla, Sudi Feng, Xinyan Liang, Shirley Dong, Xiaochun Xie, Naomi S Sta Maria, Jian-Fu Chen, Qingyi Ma, and Zhen Zhao
- Subjects
Amyloid ,Traumatic brain injury ,business.industry ,medicine.disease ,Pathogenesis ,medicine.anatomical_structure ,Gliosis ,Cerebral blood flow ,medicine ,Pericyte ,medicine.symptom ,business ,Neuroscience ,Pathological ,Neuroinflammation - Abstract
IntroductionTraumatic brain injury (TBI) is considered as the most robust environmental risk factor for Alzheimer’s disease (AD). Besides direct neuronal injury and neuroinflammation, vascular impairment is also a hallmark event of the pathological cascade after TBI. However, the vascular connection between TBI and subsequent AD pathogenesis remains underexplored.MethodsWe established a closed-head mild TBI (mTBI) model in mice with controlled cortical impact, and examined the time courses of microvascular injury, blood-brain barrier (BBB) dysfunction, gliosis and motor function impairment in wild type C57BL/6 mice. We also determined the brain clearance of β-amyloid, as well as amyloid pathology and cognitive functions after mTBI in the 5xFAD mouse model of AD.ResultsmTBI induced microvascular injury with BBB breakdown, pericyte loss and cerebral blood flow reduction in mice, which preceded gliosis. mTBI also impaired brain amyloid clearance via the vascular pathways. More importantly, mTBI accelerated amyloid pathology and cognitive impairment in the 5xFAD mice.DiscussionOur data demonstrated that microvascular injury plays a key role in the pathogenesis of AD after mTBI. Therefore, restoring vascular functions might be beneficial for patients with mTBI, and potentially reduce the risk of developing AD.
- Published
- 2020
- Full Text
- View/download PDF
33. Understanding the role of ursodeoxycholic acid and gut microbiome in non-alcoholic fatty liver disease: current evidence and perspectives
- Author
-
Qingyi Mao, Beibei Lin, Wenluo Zhang, Yu Zhang, Qian Cao, and Mengque Xu
- Subjects
ursodeoxycholic acid ,gut microbiome ,bile acid receptors ,non-alcoholic fatty liver disease ,bile acid–gut microbiome axis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, resulting in a huge medical burden worldwide. Accumulating evidence suggests that the gut microbiome and bile acids play pivotal roles during the development of NAFLD. Patients with NAFLD exhibit unique signatures of the intestinal microbiome marked by the priority of Gram-negative bacteria, decreased ratio of Firmicutes/Bacteroidetes (F/B), and increased Prevotella and Lachnospiraceae. The intestinal microbiota is involved in the metabolism of bile acids. Ursodeoxycholic acid (UDCA) is a key determinant in maintaining the dynamic communication between the host and gut microbiota. It generally shows surprising therapeutic potential in NAFLD with several mechanisms, such as improving cellular autophagy, apoptosis, and mitochondrial functions. This action is based on its direct or indirect effect, targeting the farnesoid X receptor (FXR) and various other nuclear receptors. This review aims to discuss the current studies on the involvement of the microbiome–UDCA interface in NAFLD therapy and provide prospective insights into future preventative and therapeutic approaches for NAFLD.
- Published
- 2024
- Full Text
- View/download PDF
34. C-Type Natriuretic Peptide Ameliorates Vascular Injury and Improves Neurological Outcomes in Neonatal Hypoxic-Ischemic Brain Injury in Mice
- Author
-
Lubo Zhang, Guofang Shen, Shirley Hu, Zhen Zhao, and Qingyi Ma
- Subjects
Male ,vascular protection ,Brain Edema ,Pharmacology ,Hypoxic Ischemic Encephalopathy ,Immunoglobulin G ,Mice ,Natriuretic peptide ,hypoxic-ischemic encephalopathy ,Biology (General) ,Receptor ,Cells, Cultured ,Spectroscopy ,biology ,Brain ,General Medicine ,NPR2 ,endothelial cells ,Extravasation ,Computer Science Applications ,Chemistry ,Infusions, Intraventricular ,Neuroprotective Agents ,Hypoxia-Ischemia, Brain ,Female ,Brain Infarction ,QH301-705.5 ,medicine.drug_class ,Article ,Catalysis ,Inorganic Chemistry ,Atrophy ,In vivo ,medicine ,Animals ,NPR3 ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,business.industry ,Organic Chemistry ,Natriuretic Peptide, C-Type ,Vascular System Injuries ,medicine.disease ,Animals, Newborn ,Brain Injuries ,biology.protein ,business ,C-type natriuretic peptide - Abstract
C-type natriuretic peptide (CNP) is an important vascular regulator that is present in the brain. Our previous study demonstrated the innate neuroprotectant role of CNP in the neonatal brain after hypoxic-ischemic (HI) insults. In this study, we further explored the role of CNP in cerebrovascular pathology using both in vivo and in vitro models. In a neonatal mouse HI brain injury model, we found that intracerebroventricular administration of recombinant CNP dose-dependently reduces brain infarct size. CNP significantly decreases brain edema and immunoglobulin G (IgG) extravasation into the brain tissue, suggesting a vasculoprotective effect of CNP. Moreover, in primary brain microvascular endothelial cells (BMECs), CNP dose-dependently protects BMEC survival and monolayer integrity against oxygen-glucose deprivation (OGD). The vasculoprotective effect of CNP is mediated by its innate receptors NPR2 and NPR3, in that inhibition of either NPR2 or NPR3 counteracts the protective effect of CNP on IgG leakage after HI insult and BMEC survival under OGD. Of importance, CNP significantly ameliorates brain atrophy and improves neurological deficits after HI insults. Altogether, the present study indicates that recombinant CNP exerts vascular protection in neonatal HI brain injury via its innate receptors, suggesting a potential therapeutic target for the treatment of neonatal HI brain injury.
- Published
- 2021
- Full Text
- View/download PDF
35. MicroRNA-210 suppresses glucocorticoid receptor expression in response to hypoxia in fetal rat cardiomyocytes
- Author
-
Chiranjib Dasgupta, Shannalee R. Martinez, Qingyi Ma, Xianmei Meng, and Lubo Zhang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Programmed cell death ,microRNA-210 ,Intrauterine growth restriction ,cardiomyocyte ,Biology ,03 medical and health sciences ,Glucocorticoid receptor ,Internal medicine ,glucocorticoid receptor ,medicine ,Fetus ,Gene knockdown ,hypoxia ,Hypoxia (medical) ,medicine.disease ,3. Good health ,fetal programming ,030104 developmental biology ,Endocrinology ,Oncology ,Cell culture ,medicine.symptom ,Ex vivo ,Research Paper - Abstract
// Shannalee R. Martinez 1 , Qingyi Ma 1 , Chiranjib Dasgupta 1 , Xianmei Meng 1 and Lubo Zhang 1 1 Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA Correspondence to: Lubo Zhang, email: lzhang@llu.edu Keywords: hypoxia, microRNA-210, glucocorticoid receptor, cardiomyocyte, fetal programming Received: April 04, 2017 Accepted: April 29, 2017 Published: May 11, 2017 ABSTRACT Hypoxia is a common intrauterine stressor, often resulting in intrauterine growth restriction and increased risk for cardiovascular disease later in life. The aim of this work was to test the hypothesis that microRNA-210 (miR-210) mediates the detrimental suppression of glucocorticoid receptor (GR) in response to hypoxia in fetal rat cardiomyocytes. Cardiomyocytes isolated from gestational day 21 Sprague Dawley fetal rats showed increased miR-210 levels and reduced GR abundance after exposure to ex vivo hypoxia (1% O2). In regard to mechanisms, the different contributions of hypoxia response elements (HREs) motifs in the regulation of miR-210 promoter activity and the miR-210-mediated repression of GR expression were determined in rat embryonic heart-derived myogenic cell line H9c2. Moreover, using a cell culture-based model of hypoxia-reoxygenation injury, we assessed the cytotoxic effects of GR suppression under hypoxic conditions. The results showed that hypoxia induced HIF-1α-dependent miR-210 production, as well as miR-210-mediated GR suppression, in cardiomyocytes. Furthermore, inhibition or knockdown of GR exacerbated cell death in response to hypoxia-reoxygenation injury. Altogether, the present study demonstrates that the HIF-1α-dependent miR-210-mediated suppression of GR in fetal rat cardiomyocytes increases cell death in response to hypoxia, providing novel evidence for a possible mechanistic link between fetal hypoxia and programming of ischemic-sensitive phenotype in the developing heart.
- Published
- 2017
- Full Text
- View/download PDF
36. Nutritional Stress and Fetal Epigenetics in the Brain
- Author
-
Lubo Zhang and Qingyi Ma
- Subjects
Fetus ,Physiology ,Epigenetics ,Biology - Published
- 2019
- Full Text
- View/download PDF
37. Predicting response to immunotherapy in gastric cancer via assessing perineural invasion-mediated inflammation in tumor microenvironment
- Author
-
Xunjun Li, Yiyun Wang, ZhongYa Zhai, Qingyi Mao, Dianjie Chen, Luxi Xiao, Shuai Xu, Qilin Wu, Keming Chen, Qiantong Hou, Qinglie He, Yuyang Shen, Manchun Yang, Zishan Peng, Siqing He, Xuanhui Zhou, Haoyang Tan, Shengwei Luo, Chuanfa Fang, Guoxin Li, and Tao Chen
- Subjects
Perinueral invasion(PNI) ,Gastric cancer(GC) ,Inflammatory ,Tumor microenvironment(TME) ,Neuroinflammation infiltration(NII) score system ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The perineural invasion (PNI)-mediated inflammation of the tumor microenvironment (TME) varies among gastric cancer (GC) patients and exhibits a close relationship with prognosis and immunotherapy. Assessing the neuroinflammation of TME is important in predicting the response to immunotherapy in GC patients. Methods Fifteen independent cohorts were enrolled in this study. An inflammatory score was developed and validated in GC. Based on PNI-related prognostic inflammatory signatures, patients were divided into Clusters A and B using unsupervised clustering. The characteristics of clusters and the potential regulatory mechanism of key genes were verified by RT-PCR, western-blot, immunohistochemistry and immunofluorescence in cell and tumor tissue samples.The neuroinflammation infiltration (NII) scoring system was developed based on principal component analysis (PCA) and visualized in a nomogram together with other clinical characteristics. Results Inflammatory scores were higher in GC patients with PNI compared with those without PNI (P
- Published
- 2023
- Full Text
- View/download PDF
38. Inhibition of microRNA-210 provides neuroprotection in hypoxic–ischemic brain injury in neonatal rats
- Author
-
Lubo Zhang, Fuxia Xiong, Benjamin Harding, Chiranjib Dasgupta, Nikita M. Bajwa, Richard E. Hartman, Yong Li, and Qingyi Ma
- Subjects
0301 basic medicine ,Intranasal delivery ,Encephalopathy ,Oligonucleotides ,Endogeny ,Glucocorticoid receptor ,Pharmacology ,Neuroprotection ,Article ,lcsh:RC321-571 ,03 medical and health sciences ,Receptors, Glucocorticoid ,0302 clinical medicine ,microRNA ,Animals ,Medicine ,Gene silencing ,3' Untranslated Regions ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Neonatal hypoxic–ischemic brain injury ,business.industry ,Therapeutic effect ,Brain ,medicine.disease ,Rats ,Complementary locked nucleic acid (LNA) oligonucleotides ,MicroRNAs ,Neuroprotective Agents ,030104 developmental biology ,Animals, Newborn ,Neurology ,Hypoxia-Ischemia, Brain ,Nasal administration ,business ,MicroRNA-210 ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Perinatal hypoxic–ischemic encephalopathy (HIE) is associated with high neonatal mortality and severe long-term neurologic morbidity. Yet the mechanisms of brain injury in infants with HIE remain largely elusive. The present study determined a novel mechanism of microRNA-210 (miR-210) in silencing endogenous neuroprotection and increasing hypoxic–ischemic brain injury in neonatal rats. The study further revealed a potential therapeutic effect of miR-210 inhibition using complementary locked nucleic acid oligonucleotides (miR-210-LNA) in 10-day-old neonatal rats in the Rice–Vannucci model. The underlying mechanisms were investigated with intracerebroventricular injection (i.c.v) of miR-210 mimic, miR-210-LNA, glucocorticoid receptor (GR) agonist and antagonist. Luciferase reporter gene assay was conducted for identification of miR-210 targeting GR 3′untranslated region. The results showed that the HI treatment significantly increased miR-210 levels in the brain, and miR-210 mimic significantly decreased GR protein abundance and exacerbated HI brain injury in the pups. MiR-210-LNA administration via i.c.v. 4 h after the HI insult significantly decreased brain miR-210 levels, increased GR protein abundance, reduced HI-induced neuronal death and brain infarct size, and improved long-term neurological function recovery. Of importance, the intranasal delivery of miR-210-LNA 4 h after the HI insult produced similar effects in decreasing HI-induced neonatal brain injury and improving neurological function later in life. Altogether, the present study provides evidence of a novel mechanism of miR-210 in a neonatal HI brain injury model, and suggests a potential therapeutic approach of miR-210 inhibition in the treatment of neonatal HIE.
- Published
- 2016
- Full Text
- View/download PDF
39. Antenatal hypoxia induces epigenetic repression of glucocorticoid receptor and promotes ischemic-sensitive phenotype in the developing heart
- Author
-
Zhice Xu, Shannalee R. Martinez, Qingyi Ma, Juanxiu Lv, Daliao Xiao, Minwoo Song, Fuxia Xiong, Thant Lin, Eugenia Mata-Greenwood, and Lubo Zhang
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Sp1 Transcription Factor ,Myocardial Reperfusion Injury ,Biology ,Decitabine ,Response Elements ,Article ,Epigenesis, Genetic ,Rats, Sprague-Dawley ,03 medical and health sciences ,Exon ,Receptors, Glucocorticoid ,Glucocorticoid receptor ,Pregnancy ,Internal medicine ,medicine ,Animals ,Hypoxia ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Psychological repression ,Sp1 transcription factor ,Binding Sites ,Promoter ,Exons ,DNA Methylation ,Rats ,Oxygen ,Phenotype ,030104 developmental biology ,Endocrinology ,Animals, Newborn ,Maternal Exposure ,Epigenetic Repression ,DNA methylation ,Azacitidine ,Female ,Cardiology and Cardiovascular Medicine ,Protein Binding - Abstract
Large studies in humans and animals have demonstrated a clear association of an adverse intrauterine environment with an increased risk of cardiovascular disease later in life. Yet mechanisms remain largely elusive. The present study tested the hypothesis that gestational hypoxia leads to promoter hypermethylation and epigenetic repression of the glucocorticoid receptor (GR) gene in the developing heart, resulting in increased heart susceptibility to ischemia and reperfusion injury in offspring. Hypoxic treatment of pregnant rats from day 15 to 21 of gestation resulted in a significant decrease of GR exon 14, 15, 16, and 17 transcripts, leading to down-regulation of GR mRNA and protein in the fetal heart. Functional cAMP-response elements (CREs) at −4408 and −3896 and Sp1 binding sites at −3425 and −3034 were identified at GR untranslated exon 1 promoters. Hypoxia significantly increased CpG methylation at the CREs and Sp1 binding sites and decreased transcription factor binding to GR exon 1 promoter, accounting for the repression of the GR gene in the developing heart. Of importance, treatment of newborn pups with 5-aza-2’-deoxycytidine reversed hypoxia-induced promoter methylation, restored GR expression and prevented hypoxia-mediated increase in ischemia and reperfusion injury of the heart in offspring. The findings demonstrate a novel mechanism of epigenetic repression of the GR gene in fetal stress-mediated programming of ischemic-sensitive phenotype in the heart.
- Published
- 2016
- Full Text
- View/download PDF
40. Fetal stress-mediated hypomethylation increases the brain susceptibility to hypoxic–ischemic injury in neonatal rats
- Author
-
Lubo Zhang, Qingyi Ma, Richard E. Hartman, Andre Obenaus, Yong Li, Shina Halavi, Daliao Xiao, and Katherine R. Concepcion
- Subjects
Male ,0301 basic medicine ,Pathology ,Reproductive health and childbirth ,Matrix metalloproteinase ,Hypoxia-inducible factor 1 alpha ,Rats, Sprague-Dawley ,0302 clinical medicine ,Pregnancy ,Infant Mortality ,5-aza-2 '-deoxycytidine ,Psychology ,Pediatric ,DNA methylation ,Brain ,Stroke ,Matrix Metalloproteinase 9 ,Neurology ,Hypoxia-Ischemia, Brain ,Matrix Metalloproteinase 2 ,Gestation ,Female ,Hypoxia-Inducible Factor 1 ,Disease Susceptibility ,medicine.symptom ,5-aza-2′-deoxycytidine ,medicine.medical_specialty ,Physical Injury - Accidents and Adverse Effects ,1.1 Normal biological development and functioning ,Clinical Sciences ,Biology ,alpha Subunit ,Fetal Hypoxia ,Article ,03 medical and health sciences ,Developmental Neuroscience ,Underpinning research ,In vivo ,Internal medicine ,Hypoxia-Ischemia ,medicine ,Hypoxia-inducible factor 1α ,Animals ,Hypoxic-ischemic brain injury ,Fetus ,Neurology & Neurosurgery ,Prevention ,Neurosciences ,Perinatal Period - Conditions Originating in Perinatal Period ,DNA Methylation ,Hypoxia (medical) ,Newborn ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Brain Disorders ,Rats ,030104 developmental biology ,Endocrinology ,Animals, Newborn ,Hypoxic–ischemic brain injury ,Sprague-Dawley ,030217 neurology & neurosurgery ,DNA hypomethylation - Abstract
Background and purpose Fetal hypoxia increases brain susceptibility to hypoxic–ischemic (HI) injury in neonatal rats. Yet mechanisms remain elusive. The present study tested the hypothesis that DNA hypomethylation plays a role in fetal stress-induced increase in neonatal HI brain injury. Methods Pregnant rats were exposed to hypoxia (10.5% O2) from days 15 to 21 of gestation and DNA methylation was determined in the developing brain. In addition, 5-aza-2′-deoxycytidine (5-Aza) was administered in day 7 pups brains and the HI treatment was conducted in day 10 pups. Brain injury was determined by in vivo MRI 48 h after the HI treatment and neurobehavioral function was evaluated 6 weeks after the HI treatment. Results Fetal hypoxia resulted in DNA hypomethylation in the developing brain, which persisted into 30-day old animals after birth. The treatment of neonatal brains with 5-Aza induced similar hypomethylation patterns. Of importance, the 5-Aza treatment significantly increased HI-induced brain injury and worsened neurobehavioral function recovery six weeks after the HI-treatment. In addition, 5-Aza significantly increased HIF-1α mRNA and protein abundance as well as matrix metalloproteinase (MMP)-2 and MMP-9, but decreased MMP-13 protein abundance in neonatal brains. Consistent with the 5-Aza treatment, hypoxia resulted in significantly increased expression of HIF-1α in both fetal and neonatal brains. Inhibition of HIF-1α blocked 5-Aza-mediated changes in MMPs and abrogated 5-Aza-induced increase in HI-mediated brain injury. Conclusion The results suggest that fetal stress-mediated DNA hypomethylation in the developing brain causes programming of hypoxic–ischemic sensitive phenotype in the brain and increases the susceptibility of neonatal brain to hypoxic–ischemic injury in a HIF-1α-dependent manner.
- Published
- 2016
- Full Text
- View/download PDF
41. Retraction Note: Pericyte loss influences Alzheimer-like neurodegeneration in mice.
- Author
-
Sagare, Abhay P., Bell, Robert D., Zhen Zhao, Qingyi Ma, Winkler, Ethan A., Ramanathan, Anita, and Zlokovic, Berislav V.
- Published
- 2024
- Full Text
- View/download PDF
42. The roles of astrocyte in the brain pathologies following ischemic stroke
- Author
-
Manaenko Anatol, Hongbin Han, Yixuan Zhang, Qingyi Ma, Linlin Sun, E Liu, and Junhao Yan
- Subjects
030506 rehabilitation ,Neuroscience (miscellaneous) ,Excitotoxicity ,medicine.disease_cause ,Glial scar ,Brain Ischemia ,03 medical and health sciences ,0302 clinical medicine ,Developmental and Educational Psychology ,Medicine ,Humans ,Pathological ,business.industry ,Glutamate receptor ,Neuroprotection ,Stroke ,Aquaporin 4 ,medicine.anatomical_structure ,Astrocytes ,Ischemic stroke ,Glymphatic system ,Neurology (clinical) ,0305 other medical science ,business ,Neuroscience ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Aim: In this work, we systematically explored the physiological functions of astrocytes and their roles following ischemic stroke, additionally, the potential therapy strategy targeting the astrocytes was also discussed. Methods: This work searched the PubMed database (including MEDLINE) until 14 Feb 2018, and furthermore, the studies were identified through cross-referencing and by consulting the experts in this field. Results: This study indicated that the astrocytes can not only play harmful roles following ischemic stroke through release of inflammatory factors and formation of glial scar but also have protective effects through quenching glutamate excitotoxicity and maintaining the clearance function of glymphatic system in brain. Conclusion: Owing to their important roles in physiological functions of brain and in the pathological conditions following ischemic stroke, the astrocytes might be a potential but promising therapeutic target for treating the ischemic stroke in the future.
- Published
- 2018
43. Repression of the Glucocorticoid Receptor Aggravates Acute Ischemic Brain Injuries in Adult Mice
- Author
-
Katherine R. Concepcion, Peng Zhang, Minwoo A. Song, Daliao Xiao, Lubo Zhang, Lei Huang, Yingjie Fu, Yong Li, and Qingyi Ma
- Subjects
0301 basic medicine ,Male ,Middle Cerebral Artery ,Tropomyosin receptor kinase B ,Pathogenesis ,lcsh:Chemistry ,Mice ,0302 clinical medicine ,Mineralocorticoid receptor ,Glucocorticoid receptor ,Glucocorticoid ,Neurotrophic factors ,Receptors ,2.1 Biological and endogenous factors ,Aetiology ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Spectroscopy ,BDNF/TrkB ,biology ,Cerebral infarction ,TrkB ,Brain ,Infarction, Middle Cerebral Artery ,General Medicine ,3. Good health ,Computer Science Applications ,Stroke ,GR ,Infarction ,Gene Knockdown Techniques ,Neurological ,RNA Interference ,MCAO ,Neurotrophin ,Signal Transduction ,medicine.medical_specialty ,Ischemia ,GR siRNA ,Small Interfering ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,Receptors, Glucocorticoid ,Internal medicine ,medicine ,ischemic stroke ,Genetics ,Animals ,Physical and Theoretical Chemistry ,Molecular Biology ,Inflammation ,Chemical Physics ,business.industry ,Organic Chemistry ,Neurosciences ,MR ,medicine.disease ,Brain Disorders ,030104 developmental biology ,Endocrinology ,BDNF ,lcsh:Biology (General) ,lcsh:QD1-999 ,nervous system ,biology.protein ,RNA ,Other Biological Sciences ,business ,Other Chemical Sciences ,030217 neurology & neurosurgery - Abstract
Strokes are one of the leading causes of mortality and chronic morbidity in the world, yet with only limited successful interventions available at present. Our previous studies revealed the potential role of the glucocorticoid receptor (GR) in the pathogenesis of neonatal hypoxic-ischemic encephalopathy (HIE). In the present study, we investigate the effect of GR knockdown on acute ischemic brain injuries in a model of focal cerebral ischemia induced by middle cerebral artery occlusion (MCAO) in adult male CD1 mice. GR siRNAs and the negative control were administered via intracerebroventricular (i.c.v.) injection 48 h prior to MCAO. The cerebral infarction volume and neurobehavioral deficits were determined 48 h after MCAO. RT-qPCR was employed to assess the inflammation-related gene expression profiles in the brain before and after MCAO. Western Blotting was used to evaluate the expression levels of GR, the mineralocorticoid receptor (MR) and the brain-derived neurotrophic factor/tropomyosin receptor kinase B (BDNF/TrkB) signaling. The siRNAs treatment decreased GR, but not MR, protein expression, and significantly enhanced expression levels of pro-inflammatory cytokines (IL-6, IL-1&beta, and TNF-&alpha, ) in the brain. Of interest, GR knockdown suppressed BDNF/TrkB signaling in adult mice brains. Importantly, GR siRNA pretreatment significantly increased the infarction size and exacerbated the neurobehavioral deficits induced by MCAO in comparison to the control group. Thus, the present study demonstrates the important role of GR in the regulation of the inflammatory responses and neurotrophic BDNF/TrkB signaling pathway in acute ischemic brain injuries in adult mice, revealing a new insight into the pathogenesis and therapeutic potential in acute ischemic strokes.
- Published
- 2018
44. Additional file 1: of Blood-brain barrier-associated pericytes internalize and clear aggregated amyloid-β42 by LRP1-dependent apolipoprotein E isoform-specific mechanism
- Author
-
Qingyi Ma, Zhao, Zhen, Sagare, Abhay, Yingxi Wu, Wang, Min, Owens, Nelly, Verghese, Philip, Herz, Joachim, Holtzman, David, and Zlokovic, Berislav
- Abstract
Table S1. Demographic and clinical features of human subjects used in this study. Figure S1. Aβ deposition in microvessels in AD patients and APPSw/0 mice. Figure S2. Biochemical analysis of Aβ42 aggregates. Figure S3. Cy3-Aβ42 cellular uptake in wild type mouse brain slices within 30 min. Figure S4. Pericyte coverages in Lrp1lox/lox and Lrp1lox/lox; Cspg4-Cre mice. Figure S5.. LRP1 and apoE suppression with siRNA. (DOCX 1454 kb)
- Published
- 2018
- Full Text
- View/download PDF
45. Central role for PICALM in amyloid–β blood–brain barrier transcytosis and clearance
- Author
-
Julie A. Schneider, Zhen Zhao, Qingyi Ma, Ethan A. Winkler, Anita Ramanathan, Takahiro Maeda, Ashim Ahuja, Justin K. Ichida, Carol A. Miller, Pan Kong, Matthew R. Halliday, Guojun Bu, Takahisa Kanekiyo, Sanket V Rege, Manami Maeda, Tohru Sugawara, Berislav V. Zlokovic, Donghui Zhu, Kassandra Kisler, Abhay P. Sagare, Nelly Chuqui Owens, and Gabriel Si
- Subjects
Pluripotent Stem Cells ,Endothelium ,Metabolic Clearance Rate ,media_common.quotation_subject ,Biology ,Blood–brain barrier ,Article ,PICALM ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Animals ,Homeostasis ,Humans ,Internalization ,Receptor ,030304 developmental biology ,media_common ,Cerebral Cortex ,Mice, Knockout ,0303 health sciences ,Amyloid beta-Peptides ,General Neuroscience ,Human brain ,Capillaries ,medicine.anatomical_structure ,Transcytosis ,Blood-Brain Barrier ,Monomeric Clathrin Assembly Proteins ,LDL receptor ,Endothelium, Vascular ,Neuroscience ,030217 neurology & neurosurgery - Abstract
PICALM is a highly validated genetic risk factor for Alzheimer's disease (AD). We found that reduced expression of PICALM in AD and murine brain endothelium correlated with amyloid-β (Aβ) pathology and cognitive impairment. Moreover, Picalm deficiency diminished Aβ clearance across the murine blood-brain barrier (BBB) and accelerated Aβ pathology in a manner that was reversible by endothelial PICALM re-expression. Using human brain endothelial monolayers, we found that PICALM regulated PICALM/clathrin-dependent internalization of Aβ bound to the low density lipoprotein receptor related protein-1, a key Aβ clearance receptor, and guided Aβ trafficking to Rab5 and Rab11, leading to Aβ endothelial transcytosis and clearance. PICALM levels and Aβ clearance were reduced in AD-derived endothelial monolayers, which was reversible by adenoviral-mediated PICALM transfer. Inducible pluripotent stem cell-derived human endothelial cells carrying the rs3851179 protective allele exhibited higher PICALM levels and enhanced Aβ clearance. Thus, PICALM regulates Aβ BBB transcytosis and clearance, which has implications for Aβ brain homeostasis and clearance therapy.
- Published
- 2015
46. Accelerated pericyte degeneration and blood–brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease
- Author
-
Carol A. Miller, Sanket V Rege, Zhen Zhao, Qingyi Ma, Ethan A. Winkler, Berislav V. Zlokovic, and Matthew R. Halliday
- Subjects
Male ,0301 basic medicine ,Apolipoprotein E ,Endothelium ,Apolipoprotein E4 ,Cell Count ,Blood–brain barrier ,Proinflammatory cytokine ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Humans ,Protein Isoforms ,Alleles ,Aged ,Aged, 80 and over ,Chemistry ,Neurodegeneration ,Original Articles ,medicine.disease ,LRP1 ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Blood-Brain Barrier ,Immunology ,cardiovascular system ,Female ,lipids (amino acids, peptides, and proteins) ,Endothelium, Vascular ,Neurology (clinical) ,Pericyte ,Pericytes ,Cardiology and Cardiovascular Medicine ,030217 neurology & neurosurgery ,Lipoprotein - Abstract
The blood–brain barrier (BBB) limits the entry of neurotoxic blood-derived products and cells into the brain that is required for normal neuronal functioning and information processing. Pericytes maintain the integrity of the BBB and degenerate in Alzheimer’s disease (AD). The BBB is damaged in AD, particularly in individuals carrying apolipoprotein E4 ( APOE4) gene, which is a major genetic risk factor for late-onset AD. The mechanisms underlying the BBB breakdown in AD remain, however, elusive. Here, we show accelerated pericyte degeneration in AD APOE4 carriers >AD APOE3 carriers >non-AD controls, which correlates with the magnitude of BBB breakdown to immunoglobulin G and fibrin. We also show accumulation of the proinflammatory cytokine cyclophilin A (CypA) and matrix metalloproteinase-9 (MMP-9) in pericytes and endothelial cells in AD ( APOE4 > APOE3), previously shown to lead to BBB breakdown in transgenic APOE4 mice. The levels of the apoE lipoprotein receptor, low-density lipoprotein receptor-related protein-1 (LRP1), were similarly reduced in AD APOE4 and APOE3 carriers. Our data suggest that APOE4 leads to accelerated pericyte loss and enhanced activation of LRP1-dependent CypA–MMP-9 BBB-degrading pathway in pericytes and endothelial cells, which can mediate a greater BBB damage in AD APOE4 compared with AD APOE3 carriers.
- Published
- 2015
- Full Text
- View/download PDF
47. Gestational hypoxia and epigenetic programming of brain development disorders
- Author
-
Lubo Zhang, Fuxia Xiong, and Qingyi Ma
- Subjects
Pharmacology ,Genetics ,Brain Diseases ,Fetus ,Offspring ,Hypoxia (medical) ,Biology ,Fetal Hypoxia ,Bioinformatics ,Article ,Epigenesis, Genetic ,Epigenetic programming ,Drug Discovery ,DNA methylation ,microRNA ,medicine ,Animals ,Humans ,Gestation ,Epigenetics ,medicine.symptom - Abstract
Adverse environmental conditions faced by an individual early during its life, such as gestational hypoxia, can have a profound influence on the risk of diseases, such as neurological disorders, in later life. Clinical and preclinical studies suggest that epigenetic programming of gene expression patterns in response to maternal stress have a crucial role in the fetal origins of neurological diseases. Herein, we summarize recent studies regarding the role of epigenetic mechanisms in the developmental programming of neurological diseases in offspring, primarily focusing on DNA methylation/demethylation and miRNAs. Such information could increase our understanding of the fetal origins of adult diseases and help develop effective prevention and intervention against neurological diseases.
- Published
- 2014
- Full Text
- View/download PDF
48. C-type natriuretic peptide functions as an innate neuroprotectant in neonatal hypoxic-ischemic brain injury in mouse via natriuretic peptide receptor 2
- Author
-
Qingyi Ma and Lubo Zhang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.drug_class ,Endogeny ,Brain damage ,Neuroprotection ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Developmental Neuroscience ,In vivo ,Internal medicine ,Natriuretic peptide ,Medicine ,Animals ,Receptor ,business.industry ,Antagonist ,Natriuretic Peptide, C-Type ,NPR2 ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Neurology ,nervous system ,Animals, Newborn ,Hypoxia-Ischemia, Brain ,medicine.symptom ,business ,Receptors, Atrial Natriuretic Factor ,030217 neurology & neurosurgery - Abstract
Neonatal hypoxia-ischemia (HI) is the most common cause of brain injury in neonates, which leads to high neonatal mortality and severe neurological morbidity in later life (Vannucci, 2000; Volpe, 2001). Yet the molecular mechanisms of neuronal death and brain damage induced by neonatal HI remain largely elusive. Herein, using both in vivo and in vitro models, we determine an endogenous neuroprotectant role of c-type natriuretic peptide (CNP) in preserving neuronal survival after HI brain injury in mouse pups. Postnatal day 7 (P7) mouse pups with CNP deficiency (Nppc(lbab/lbab)) exhibit increased brain infarct size and worsened long-term locomotor function after neonatal HI compared with wildtype control (Nppc(+/+)). In isolated primary cortical neurons, recombinant CNP dose-dependently protects primary neurons from oxygen-glucose deprivation (OGD) insult. This neuroprotective effect appears to be mediated through its cognate natriuretic peptide receptor 2 (NPR2), in that antagonization of NPR2, but not NPR3, exacerbates neuronal death and counteracts the protective effect of CNP on primary neurons exposed to OGD insult. Immunoblot and confocal microscopy demonstrate the abundant expression of NPR2 in neurons of the neonatal brain and in isolated primary cortical neurons as well. Moreover, similar to CNP deficiency, administration of NPR2 antagonist P19 via intracerebroventricular injection prior to HI results in exacerbated neuronal death and brain injury after HI. Altogether, the present study indicates that CNP and its cognate receptor NPR2 mainly expressed in neurons represent an innate neuroprotective mechanism in neonatal HI brain injury.
- Published
- 2017
49. Inhibition of microRNA-210 suppresses pro-inflammatory response and reduces acute brain injury of ischemic stroke in mice
- Author
-
Yong Li, Qingyi Ma, Lubo Zhang, Bo Li, and Lei Huang
- Subjects
0301 basic medicine ,Male ,Chemokine ,RNA, Untranslated ,Ischemia ,CCL3 ,Infarction ,Inflammation ,Brain damage ,Pharmacology ,CCL2 ,Article ,Brain Ischemia ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Developmental Neuroscience ,medicine ,Animals ,cardiovascular diseases ,Injections, Intraventricular ,biology ,Cerebral infarction ,business.industry ,medicine.disease ,Mice, Inbred C57BL ,Stroke ,MicroRNAs ,030104 developmental biology ,Neurology ,Anesthesia ,Brain Injuries ,biology.protein ,medicine.symptom ,Inflammation Mediators ,business ,030217 neurology & neurosurgery - Abstract
Stroke is a leading cause of mortality and chronic neurologic disability. Yet, the successful treatment remains limited. In this study, we investigated the efficacy and the mechanism of a novel treatment, microRNA-210 (miR-210) inhibition, in protecting acute ischemic brain injury in adult mice. Focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in adult male C57BL/6 mice. MiR-210-LNA (miR-210 inhibitor) or the negative control was administered via intracerebroventricular injection 24 h prior or 4 h after MCAO. Cerebral infarction volume and behavioral deficits were determined 48 h after MCAO. The expression of inflammation-related genes and infiltration/activation of various immune cells in the brain were assessed by RT-qPCR, flow cytometry, and immunohistochemistry. Acute ischemic stroke significantly increased miR-210 levels in the brain, which was abolished by miR-210-LNA administered prior to MCAO. Pre- and post-MCAO treatments with miR-210-LNA significantly decreased cerebral infarction and ameliorated behavioral deficits induced by MCAO. Long-term behavioral recovery was also improved by miR-210-LNA post-treatment. At the same time, inhibition of miR-210 significantly reduced the expression of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and chemokines (CCL2 and CCL3), but had no significant effect on anti-inflammatory factors (TGF-β and IL-10). In addition, MCAO-induced macrophage infiltration and microglial activation in the brain were inhibited by the miR-210-LNA treatment. In summary, inhibition of miR-210 suppresses pro-inflammatory response and reduces brain damage in the acute phase of ischemic stroke, providing new insight in molecular basis of a novel therapeutic strategy of miR-210 inhibition in the treatment of acute ischemic stroke.
- Published
- 2017
50. MicroRNA-210 Suppresses Junction Proteins and Disrupts Blood-Brain Barrier Integrity in Neonatal Rat Hypoxic-Ischemic Brain Injury
- Author
-
Lei Huang, Chiranjib Dasgupta, Qingyi Ma, Yong Li, and Lubo Zhang
- Subjects
0301 basic medicine ,Male ,Pathology ,blood-brain barrier integrity ,Brain Edema ,Neurodegenerative ,Occludin ,lcsh:Chemistry ,0302 clinical medicine ,2.1 Biological and endogenous factors ,neonatal hypoxic-ischemic brain injury ,microRNA-210 ,cerebral edema ,Aetiology ,lcsh:QH301-705.5 ,Spectroscopy ,beta Catenin ,Pediatric ,Tight junction ,Brain ,General Medicine ,3. Good health ,Computer Science Applications ,Stroke ,medicine.anatomical_structure ,Intercellular Junctions ,Blood-Brain Barrier ,Hypoxia-Ischemia, Brain ,Neurological ,RNA Interference ,Female ,Biotechnology ,medicine.medical_specialty ,Encephalopathy ,Biology ,Blood–brain barrier ,Neuroprotection ,Article ,Catalysis ,Permeability ,Cerebral edema ,Inorganic Chemistry ,Adherens junction ,03 medical and health sciences ,Internal medicine ,Parenchyma ,Hypoxia-Ischemia ,medicine ,Genetics ,Animals ,Physical and Theoretical Chemistry ,Molecular Biology ,Chemical Physics ,Animal ,Organic Chemistry ,Neurosciences ,Perinatal Period - Conditions Originating in Perinatal Period ,medicine.disease ,Newborn ,Brain Disorders ,Rats ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,Endocrinology ,Good Health and Well Being ,Animals, Newborn ,lcsh:Biology (General) ,lcsh:QD1-999 ,Gene Expression Regulation ,Immunoglobulin G ,Disease Models ,Other Biological Sciences ,Other Chemical Sciences ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Cerebral edema, primarily caused by disruption of the blood-brain barrier (BBB), is one of the serious complications associated with brain injury in neonatal hypoxic-ischemic encephalopathy (HIE). Our recent study demonstrated that the hypoxic-ischemic (HI) treatment significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of miR-210 provided neuroprotection in neonatal HI brain injury. The present study aims to determine the role of miR-210 in the regulation of BBB integrity in the developing brain. miR-210 mimic was administered via intracerebroventricular injection (i.c.v.) into the brain of rat pups. Forty-eight hours after the injection, a modified Rice-Vannucci model was conducted to produce HI brain injury. Post-assays included cerebral edema analysis, western blotting, and immunofluorescence staining for serum immunoglobulin G (IgG) leakage. The results showed that miR-210 mimic exacerbated cerebral edema and IgG leakage into the brain parenchyma. In contrast, inhibition of miR-210 with its complementary locked nucleic acid oligonucleotides (miR-210-LNA) significantly reduced cerebral edema and IgG leakage. These findings suggest that miR-210 negatively regulates BBB integrity i n the neonatal brain. Mechanistically, the seed sequences of miR-210 were identified complementary to the 3' untranslated region (3' UTR) of the mRNA transcripts of tight junction protein occludin and adherens junction protein β-catenin, indicating downstream targets of miR-210. This was further validated by in vivo data showing that miR-210 mimic significantly reduced the expression of these junction proteins in rat pup brains. Of importance, miR-210-LNA preserved the expression of junction proteins occludin and β-catenin from neonatal HI insult. Altogether, the present study reveals a novel mechanism of miR-210 in impairing BBB integrity that contributes to cerebral edema formation after neonatal HI insult, and provides new insights in miR-210-LNA mediated neuroprotection in neonatal HI brain injury.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.