13 results on '"Yanghao Hou"'
Search Results
2. A narrative review of what the neuropathologist needs to tell the clinician in neuro-oncology practice concerning WHO CNS5
- Author
-
Yanghao Hou and Felix Sahm
- Subjects
central nervous system tumors ,glioma ,methylation ,neuro-oncology ,neuropathology ,next-generation sequencing ,tumor classification ,world health organization ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The fifth edition of the World Health Organization Classification of Tumors of the Central Nervous System (WHO CNS5) was released in late 2021. The adoption of new tumor nomenclature, grading criteria, terminology, tumor types, and novel diagnostic approaches, including methylation profiling, will benefit the precise diagnosis of CNS tumors, facilitate optimal patient care and improve diagnostic reproducibility with higher clinical relevance. However, the gap between rapid updates in tumor classification and the clinical management of patients requires frequent and up-to-date communications between neuropathologists and clinicians. This review aims to provide an overview of WHO CNS5, focused on the changes that are most pertinent to the clinical care of patients. Forming concrete ideas for neuropathologists that are necessary to express to clinicians, for a better understanding of how the patient may benefit from the new classification.
- Published
- 2022
- Full Text
- View/download PDF
3. Extraventricular Neurocytoma: a Case Report and Literature Review
- Author
-
Liping Wang, Xiaoxiao Dai, Yifang Ping, Ping Chen, Yanming Chen, Shan Cheng, Yongdong Li, Suwen Li, Haoran Li, Jiaqi Yuan, Yanghao Hou, and Jun Dong
- Abstract
Extraventricular neurocytoma (EVN) is a rare intracranial tumor. The most common locations of EVN were frontal lobe and cerebellum, followed by temporal lobe. Heterogeneous contrast enhancement, cystic changes, perilesional edema and calcification were a few distinct imaging characteristics. EVN is a primary tumor with either glial or neuronal differentiation or potential for atypical changes. The lack of characteristic manifestations in histopathology makes EVN difficult to distinguish from other low-grade neuronal and mixed neuronal-glial tumors. Herein, we describe a case of frontal extraventricular neurocytoma in a 37-year-old male presenting with dizzy and review the literature on this rare tumor. Pathological examination reveals that the tumor mainly composed of oligodendroglia-like cells, with expression of synaptophysin (SYN) and Olig2 (oligodendrocyte transcription factor 2), and microcalcifications. Methylation profile and t-SNE analysis show our sample cluster with the other two reference EVNs. Next-generation sequencing (NGS) shows FGFR1-TACC1 fusion. We report a case of EVN with expression of Olig2, FGFR1-TACC1 fusion, methylation of MGMT promoter, and discuss our experience along with a review of the published literature.
- Published
- 2022
4. Mosaic trisomy of chromosome 1q in human brain tissue associates with unilateral polymicrogyria, very early-onset focal epilepsy, and severe developmental delay
- Author
-
Karl Rössler, Samir Jabari, Wim G.M. Spliet, Friedrich G. Woermann, Manfred Kudernatsch, Angelika Mühlebner, Hajo M. Hamer, Damian Stichel, Yanghao Hou, Ingmar Blümcke, Andrey Korshunov, Tom Pieper, Katja Kobow, Andreas von Deimling, Thilo Kalbhenn, Felix Sahm, Roland Coras, Martha Feucht, Tilman Polster, Pathology, APH - Aging & Later Life, APH - Mental Health, and ANS - Cellular & Molecular Mechanisms
- Subjects
0301 basic medicine ,Male ,Hemimegalencephaly ,Pathology ,medicine.medical_specialty ,Drug Resistant Epilepsy ,DNA Copy Number Variations ,Unilateral polymicrogyria ,Biology ,Chromosomes ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Epilepsy ,0302 clinical medicine ,Seizures ,Gene duplication ,medicine ,Polymicrogyria ,Humans ,Copy-number variation ,ddc:610 ,Original Paper ,ID ,Cortical malformation ,Copy number variation ,Brain ,Chromosome 1 ,Cortical dysplasia ,medicine.disease ,Brain development ,Malformations of Cortical Development ,030104 developmental biology ,DNA methylation ,Female ,Neurology (clinical) ,Trisomy ,030217 neurology & neurosurgery - Abstract
Polymicrogyria (PMG) is a developmental cortical malformation characterized by an excess of small and frustrane gyration and abnormal cortical lamination. PMG frequently associates with seizures. The molecular pathomechanisms underlying PMG development are not yet understood. About 40 genes have been associated with PMG, and small copy number variations have also been described in selected patients. We recently provided evidence that epilepsy-associated structural brain lesions can be classified based on genomic DNA methylation patterns. Here, we analyzed 26 PMG patients employing array-based DNA methylation profiling on formalin-fixed paraffin-embedded material. A series of 62 well-characterized non-PMG cortical malformations (focal cortical dysplasia type 2a/b and hemimegalencephaly), temporal lobe epilepsy, and non-epilepsy autopsy controls was used as reference cohort. Unsupervised dimensionality reduction and hierarchical cluster analysis of DNA methylation profiles showed that PMG formed a distinct DNA methylation class. Copy number profiling from DNA methylation data identified a uniform duplication spanning the entire long arm of chromosome 1 in 7 out of 26 PMG patients, which was verified by additional fluorescence in situ hybridization analysis. In respective cases, about 50% of nuclei in the center of the PMG lesion were 1q triploid. No chromosomal imbalance was seen in adjacent, architecturally normal-appearing tissue indicating mosaicism. Clinically, PMG 1q patients presented with a unilateral frontal or hemispheric PMG without hemimegalencephaly, a severe form of intractable epilepsy with seizure onset in the first months of life, and severe developmental delay. Our results show that PMG can be classified among other structural brain lesions according to their DNA methylation profile. One subset of PMG with distinct clinical features exhibits a duplication of chromosomal arm 1q. Electronic supplementary material The online version of this article (10.1007/s00401-020-02228-5) contains supplementary material, which is available to authorized users.
- Published
- 2020
5. LGG-13. PAPILLARY GLIONEURONAL TUMOR (PGNT) EXHIBITS A CHARACTERISTIC METHYLATION PROFILE AND MANDATORY FUSIONS INVOLVING PRKCA
- Author
-
David T.W. Jones, Felix Sahm, Luca Bertero, Stefan M. Pfister, Yanghao Hou, Jorge Pinheiro, and Andreas von Deimling
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Pilocytic astrocytoma ,Dysembryoplastic Neuroepithelial Tumor ,Low Grade Glioma ,Methylation ,Biology ,medicine.disease ,Oncology ,medicine ,Papillary glioneuronal tumor ,Neurology (clinical) ,Notch1 gene - Abstract
Papillary glioneuronal tumor (PGNT) predominantly arises in children, adolescents and young adults. Its diagnosis poses a major diagnostic challenge. Recently, SLC44A1-PRKCA fusions have been described in PGNT. We subjected 28 brain tumors from different institutions histologically diagnosed as PGNT to molecular analysis and morphological analysis. Array-based methylation analysis revealed that 17/28 tumors exhibited methylation profiles typical for other tumor entities, mostly dysembryoplastic neuroepithelial tumor and hemispheric pilocytic astrocytoma. Conversely, 11/28 tumors exhibited a unique profile thus constituting a distinct methylation class PGNT. By screening the extended Heidelberg cohort containing over 25000 CNS tumors, we identified three additional tumors belonging to this methylation cluster, but originally histologically diagnosed otherwise. RNA sequencing for the detection of SLC44A1-PRKCA fusions could be performed on 19 of the tumors, 10 of them belonging to the methylation class PGNT. In two additional cases, SLC44A1-PRKCA fusions were confirmed by FISH. We detected fusions involving PRKCA in all of the cases of this methylation class with material available for analyses: the canonical SLC44A1-PRKCA fusion was observed in 11/12 tumors, while the remaining case exhibited a NOTCH1-PRKCA fusion. Neither of the fusions was found in the tumors belonging to other methylation classes. Our results point towards a high misclassification rate of the morphological diagnosis PGNT and clearly demonstrate the necessity of molecular analyses. PRKCA fusions are highly diagnostic for PGNT and detection by RNA sequencing allows identification of rare fusion partners. Methylation analysis recognizes a unique methylation class PGNT irrespective of the nature of the PRKCA Fusion.
- Published
- 2019
6. Papillary glioneuronal tumor (PGNT) exhibits a characteristic methylation profile and fusions involving PRKCA
- Author
-
Marco Prinz, Andrey Korshunov, Stefan Pusch, Daniel Schrimpf, Karin de Stricker, Leonille Schweizer, Sebastian Brandner, Werner Paulus, Mélanie Pagès, David T.W. Jones, Ingmar Blümcke, Guido Reifenberger, Andreas von Deimling, Stefan M. Pfister, Jürgen Hench, Damian Stichel, Bianca Pollo, Azadeh Ebrahimi, Pascale Varlet, Francisco Fernández-Klett, Felix Sahm, Jorge Pinheiro, David Scheie, David Capper, Karl H. Plate, Belen Casalini, Ulrich Schüller, Jochen Meier, Luca Bertero, Annekathrin Reinhardt, Yanghao Hou, Torsten Pietsch, Wolfgang Wick, David E. Reuss, Philipp Sievers, Annika K. Wefers, Christian Hartmann, Christian Koelsche, Stephan Frank, and Andreas Unterberg
- Subjects
Adult ,Male ,0301 basic medicine ,Site-Specific DNA-Methyltransferase (Adenine-Specific) ,SLC44A1 ,Protein Kinase C-alpha ,Adolescent ,Organic Cation Transport Proteins ,PRKCA ,Papillary glioneuronal tumor ,Brain tumor ,Biology ,Pathology and Forensic Medicine ,Cohort Studies ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,NOTCH1 ,Antigens, CD ,Methylation analysis ,Biomarkers, Tumor ,medicine ,Humans ,CNS TUMORS ,Child ,DNA methylation ,RNA sequencing ,Pilocytic astrocytoma ,Brain Neoplasms ,Dysembryoplastic Neuroepithelial Tumor ,Brain ,Methylation ,Middle Aged ,medicine.disease ,Neoplasms, Neuroepithelial ,030104 developmental biology ,Cancer research ,Female ,Neurology (clinical) ,Gene Fusion ,030217 neurology & neurosurgery - Abstract
Papillary glioneuronal tumor (PGNT) is a WHO-defined brain tumor entity that poses a major diagnostic challenge. Recently, SLC44A1-PRKCA fusions have been described in PGNT. We subjected 28 brain tumors from different institutions histologically diagnosed as PGNT to molecular and morphological analysis. Array-based methylation analysis revealed that 17/28 tumors exhibited methylation profiles typical for other tumor entities, mostly dysembryoplastic neuroepithelial tumor and hemispheric pilocytic astrocytoma. Conversely, 11/28 tumors exhibited a unique profile, thus constituting a distinct methylation class PGNT. By screening the extended Heidelberg cohort containing over 25,000 CNS tumors, we identified three additional tumors belonging to this methylation cluster but originally histologically diagnosed otherwise. RNA sequencing for the detection of SLC44A1-PRKCA fusions could be performed on 19 of the tumors, 10 of them belonging to the methylation class PGNT. In two additional cases, SLC44A1-PRKCA fusions were confirmed by FISH. We detected fusions involving PRKCA in all cases of this methylation class with material available for analyses: the canonical SLC44A1-PRKCA fusion was observed in 11/12 tumors, while the remaining case exhibited a NOTCH1-PRKCA fusion. Neither of the fusions was found in the tumors belonging to other methylation classes. Our results point towards a high misclassification rate of the morphological diagnosis PGNT and clearly demonstrate the necessity of molecular analyses. PRKCA fusions are highly diagnostic for PGNT, and detection by RNA sequencing enables the identification of rare fusion partners. Methylation analysis recognizes a unique methylation class PGNT irrespective of the nature of the PRKCA fusion.
- Published
- 2019
7. Primary intracranial spindle cell sarcoma with rhabdomyosarcoma-like features share a highly distinct methylation profile and DICER1 mutations
- Author
-
Andreas von Deimling, Javier Alonso, Andrey Korshunov, Felix Sahm, Luca Bertero, Daniel Schrimpf, Gunhild Mechtersheimer, Uta Flucke, Daniel Baumhoer, Jonathan Serrano, Yanghao Hou, Oscar M. Tirado, Jaume Mora, Juan Luis Garcia Leon, David T.W. Jones, Manfred Gessler, Dominik Sturm, Christian Vokuhl, Wolfgang Brück, Rosdali Y. Diaz Coronado, Stefan Rutkowski, Manel Esteller, Ulrich Schüller, Sandro Casavilca Zambrano, Till Milde, Jonas Ecker, Iver Petersen, Matija Snuderl, Stefan M. Pfister, Xavier Garcia del Muro, David E. Reuss, Martin Mynarek, Christian Koelsche, German Cancer Aid, Friedberg Charitable Foundation, Sohn Conference Foundation, Fördergemeinschaft Kinderkrebs-Zentrum Hamburg, Damp Foundation, and Damp Foundation (to M.M.).
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,DNA-methylation profiling ,Male ,Ribonuclease III ,Pathology ,Pleuropulmonary blastoma ,DICER1 ,medicine.disease_cause ,DEAD-box RNA Helicases ,0302 clinical medicine ,Rhabdomyosarcoma, Embryonal ,TP53 ,Rhabdomyosarcoma ,Sarcoma ,Cystic nephroma ,MAPK ,030220 oncology & carcinogenesis ,NGS ,EPIC array ,Female ,KRAS ,CNS ,medicine.medical_specialty ,Biology ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,All institutes and research themes of the Radboud University Medical Center ,medicine ,Humans ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Retrospective Studies ,DNA Methylation ,medicine.disease ,030104 developmental biology ,2734 ,Neurology (clinical) ,Mutation ,Embryonal rhabdomyosarcoma ,Spindle cell sarcoma - Abstract
Patients with DICER1 predisposition syndrome have an increased risk to develop pleuropulmonary blastoma, cystic nephroma, embryonal rhabdomyosarcoma, and several other rare tumor entities. In this study, we identified 22 primary intracranial sarcomas, including 18 in pediatric patients, with a distinct methylation signature detected by array-based DNA-methylation profiling. In addition, two uterine rhabdomyosarcomas sharing identical features were identified. Gene panel sequencing of the 22 intracranial sarcomas revealed the almost unifying feature of DICER1 hotspot mutations (21/22; 95%) and a high frequency of co-occurring TP53 mutations (12/22; 55%). In addition, 17/22 (77%) sarcomas exhibited alterations in the mitogen-activated protein kinase pathway, most frequently affecting the mutational hotspots of KRAS (8/22; 36%) and mutations or deletions of NF1 (7/22; 32%), followed by mutations of FGFR4 (2/22; 9%), NRAS (2/22; 9%), and amplification of EGFR (1/22; 5%). A germline DICER1 mutation was detected in two of five cases with constitutional DNA available. Notably, none of the patients showed evidence of a cancer-related syndrome at the time of diagnosis. In contrast to the genetic findings, the morphological features of these tumors were less distinctive, although rhabdomyoblasts or rhabdomyoblast-like cells could retrospectively be detected in all cases. The identified combination of genetic events indicates a relationship between the intracranial tumors analyzed and DICER1 predisposition syndrome-associated sarcomas such as embryonal rhabdomyosarcoma or the recently described group of anaplastic sarcomas of the kidney. However, the intracranial tumors in our series were initially interpreted to represent various tumor types, but rhabdomyosarcoma was not among the typical differential diagnoses considered. Given the rarity of intracranial sarcomas, this molecularly clearly defined group comprises a considerable fraction thereof. We therefore propose the designation "spindle cell sarcoma with rhabdomyosarcoma-like features, DICER1 mutant" for this intriguing group.
- Published
- 2018
8. FGFR1:TACC1 fusion is a frequent event in molecularly defined extraventricular neurocytoma
- Author
-
Roland Coras, Elisabeth J. Rushing, Rudi Beschorner, Kristian W. Pajtler, David E. Reuss, Walter Stummer, Daniel Schrimpf, Andreas von Deimling, Stefan M. Pfister, Caterina Giannini, Christian Hagel, Felice Giangaspero, Philipp Sievers, Uta Schick, Christel Herold-Mende, Annika K. Wefers, Azadeh Ebrahimi, Patricia Kohlhof, Kristin Huang, Andrey Korshunov, Ori Staszewski, Francesca Diomedi-Camassei, David T.W. Jones, Christian Koelsche, Guido Reifenberger, Felix Sahm, Yanghao Hou, Damian Stichel, Annekathrin Reinhardt, Christian Hartmann, Martin Hasselblatt, Kathy Keyvani, Sievers P., Stichel D., Schrimpf D., Sahm F., Koelsche C., Reuss D.E., Wefers A.K., Reinhardt A., Huang K., Ebrahimi A., Hou Y., Pajtler K.W., Pfister S.M., Hasselblatt M., Stummer W., Schick U., Hartmann C., Hagel C., Staszewski O., Reifenberger G., Beschorner R., Coras R., Keyvani K., Kohlhof P., Diomedi-Camassei F., Herold-Mende C., Giangaspero F., Rushing E., Giannini C., Korshunov A., Jones D.T.W., von Deimling A., University of Zurich, and von Deimling, Andreas
- Subjects
Fetal Proteins ,Male ,0301 basic medicine ,Pathology ,Oncogene Proteins, Fusion ,2804 Cellular and Molecular Neuroscience ,Medizin ,Kaplan-Meier Estimate ,DNA methylation profile ,Histones ,0302 clinical medicine ,Retrospective Studie ,Neurocytoma ,Nuclear Protein ,Brain Neoplasms ,FGFR ,Nuclear Proteins ,Methylation ,Isocitrate Dehydrogenase ,Histone ,2728 Neurology (clinical) ,Extraventricular neurocytoma ,Molecular classification ,DNA methylation ,Female ,Microtubule-Associated Proteins ,Human ,medicine.medical_specialty ,10208 Institute of Neuropathology ,Clinical Neurology ,Brain tumor ,Copy number analysis ,610 Medicine & health ,Biology ,Pathology and Forensic Medicine ,Brain Neoplasm ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Text mining ,Fetal Protein ,Parenchyma ,medicine ,Central neurocytoma ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Epigenetics ,Fusion ,Retrospective Studies ,business.industry ,Microtubule-Associated Protein ,DNA Methylation ,medicine.disease ,FGFR1–TACC1 ,2734 Pathology and Forensic Medicine ,Ki-67 Antigen ,030104 developmental biology ,570 Life sciences ,biology ,brain tumor ,extraventricular neurocytoma ,fusion ,molecular classification ,Neurology (clinical) ,Transcriptome ,business ,030217 neurology & neurosurgery - Abstract
Extraventricular neurocytoma (EVN) is a rare primary brain tumor occurring in brain parenchyma outside the ventricular system. Histopathological characteristics resemble those of central neurocytoma but exhibit a wider morphologic spectrum. Accurate diagnosis of these histologically heterogeneous tumors is often challenging because of the overlapping morphological features and the lack of defining molecular markers. Here, we explored the molecular landscape of 40 tumors diagnosed histologically as EVN by investigating copy number profiles and DNA methylation array data. DNA methylation profiles were compared with those of relevant differential diagnoses of EVN and with a broader spectrum of diverse brain tumor entities. Based on this, our tumor cohort segregated into different groups. While a large fraction (n = 22) formed a separate epigenetic group clearly distinct from established DNA methylation profiles of other entities, a subset (n = 14) of histologically diagnosed EVN grouped with clusters of other defined entities. Three cases formed a small group close to but separated from the epigenetically distinct EVN cases, and one sample clustered with non-neoplastic brain tissue. Four additional samples originally diagnosed otherwise were found to molecularly resemble EVN. Thus, our results highlight a distinct DNA methylation pattern for the majority of tumors diagnosed as EVN, but also indicate that approximately onethird of morphological diagnoses of EVN epigenetically correspond to other brain tumor entities. Copy number analysis and confirmation through RNA sequencing revealed FGFR1–TACC1 fusion as a distinctive, recurrent feature within the EVN methylation group (60%), in addition to a small number of other FGFR rearrangements (13%). In conclusion, our data demonstrate a specific epigenetic signature of EVN suitable for characterization of these tumors as a molecularly distinct entity, and reveal a high frequency of potentially druggable FGFR pathway activation in this tumor group.
- Published
- 2018
9. Resveratrol alleviates cerebral ischemia/reperfusion injury in rats by inhibiting NLRP3 inflammasome activation through Sirt1-dependent autophagy induction
- Author
-
Lingyu Li, Qi He, Zhenyu Li, Jing Zhao, Yueting Wang, and Yanghao Hou
- Subjects
0301 basic medicine ,Agonist ,Male ,endocrine system diseases ,medicine.drug_class ,Inflammasomes ,Immunology ,Ischemia ,Inflammation ,Resveratrol ,Pharmacology ,Neuroprotection ,Brain Ischemia ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Sirtuin 1 ,NLR Family, Pyrin Domain-Containing 3 Protein ,Stilbenes ,medicine ,Autophagy ,Immunology and Allergy ,Animals ,Humans ,business.industry ,Anti-Inflammatory Agents, Non-Steroidal ,food and beverages ,Inflammasome ,Cerebral Arteries ,medicine.disease ,Rats ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Reperfusion Injury ,medicine.symptom ,business ,Reperfusion injury ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Resveratrol has been reported to protect against cerebral ischemia/reperfusion (I/R) injury in rats, but the underlying mechanism is unclear. In the current study, we examined whether resveratrol ameliorates cerebral I/R injury by inhibiting NLRP3 inflammasome-derived inflammation and whether autophagy is involved in this process. In addition, we explored the role of Sirt1 in resveratrol-mediated protective effects. To answer these questions, healthy male Sprague-Dawley rats were exposed to middle cerebral artery occlusion for 1 h followed by 24 h reperfusion. We found that cerebral I/R increased levels of activated NLRP3 inflammasome, caspase-1, IL-1β, and IL-18 and enhanced autophagy activity (ratio of LC3B-II/LC3B-I and p62/SQSTM1). Treatment with resveratrol, a specific Sirt1 agonist, attenuated I/R-induced NLRP3 inflammasome-derived inflammation but upregulated autophagy. Furthermore, resveratrol treatment clearly reduced cerebral infarct volume, decreased brain water content, and improved neurological scores. In addition, inhibition of autophagy using 3-MA intracerebroventricular injection blocked the inhibitory effect of resveratrol on NLRP3 inflammasome activation. Finally, Sirt1 knockdown with siRNA significantly blocked resveratrol-induced enhancement of autophagy activity and suppression of NLRP3 inflammasome activation. In conclusion, our results demonstrate that resveratrol protects against cerebral I/R injury by inhibiting NLRP3 inflammasome activation through Sirt1-dependent autophagy activity.
- Published
- 2017
10. Nrf2 inhibits NLRP3 inflammasome activation through regulating Trx1/TXNIP complex in cerebral ischemia reperfusion injury
- Author
-
Hui Xie, Jing Zhao, Yueting Wang, Lingyu Li, Yong Zhao, Yanghao Hou, and Qi He
- Subjects
0301 basic medicine ,Male ,Thioredoxin-Interacting Protein ,Inflammasomes ,NF-E2-Related Factor 2 ,Interleukin-1beta ,Ischemia ,Caspase 1 ,Inflammation ,Cell Cycle Proteins ,Pharmacology ,Biology ,environment and public health ,Antioxidants ,Brain Ischemia ,Brain ischemia ,Rats, Sprague-Dawley ,03 medical and health sciences ,Behavioral Neuroscience ,Thioredoxins ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Interleukin-18 ,Inflammasome ,Infarction, Middle Cerebral Artery ,respiratory system ,medicine.disease ,Rats ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,Reperfusion Injury ,Immunology ,medicine.symptom ,Carrier Proteins ,Reperfusion injury ,TXNIP ,medicine.drug - Abstract
The nod-like receptor protein 3 (NLRP3) inflammasome has a critical role in inflammation damage in ischemic injury, and the activation of the inflammasome is closely related to the interaction with thioredoxin interacting protein (TXNIP), which dissociates from the thioredoxin1 (Trx1)/TXNIP complex under oxidative stress. However, the negative regulator of NLRP3 inflammasome activation has not been fully investigated. Nuclear factor erythroid 2-related factor 2 (Nrf2) takes on a critical part in the antioxidant stress system, that controls the driven genes of antioxidant response element (ARE). Activate Nrf2 could inhibit the activation of NLRP3 inflammasome in acute liver injury and severe lupus nephritis. We aimed to explore the protective effect of Nrf2 in inhibiting the NLPR3 inflammasome formulation through the Trx1/TXNIP complex in cerebral ischemia reperfusion (cerebral I/R) injury. Middle cerebral artery occlusion/reperfusion (MCAO/R) model was used to imitate ischemic insult. Nrf2 was activated by tert-butylhydroquinone (tBHQ) intraperitoneally (i.p.) injection (16.7mg/kg), Nrf2,Trx1 and NLRP3 siRNAs were infused into the left paracele (12μl per rat), protein and mRNA levels were assessed by Western blot, qRT-PCR. ELISA was used for IL-1β and IL-18 activity measurements. After upregulating Nrf2, the expression of TXNIP in cytoplasm, NLRP3 inflammasome, and downstream factors caspase-1, IL-18, and IL-1β were significantly reduced, and Nrf2 knockdown yielded the opposite results. Trx1 knockdown produced the same effect of Nrf2 inhibition and the protective effect of Nrf2 was mostly abolished. Our results suggested that Nrf2 acted as a protective regulator against NLRP3 inflammasome activation by regulating the Trx1/TXNIP complex, which could possibly represent an innovative insight into the treatment of ischemia and reperfusion injury.
- Published
- 2017
11. Sestrin2 Silencing Exacerbates Cerebral Ischemia/Reperfusion Injury by Decreasing Mitochondrial Biogenesis through the AMPK/PGC-1α Pathway in Rats
- Author
-
Lina Xiao, Jin Zhu, Shanshan Yu, Yanghao Hou, Yixin Li, Jing Zhao, Lingyu Li, Qi He, Jingxian Wu, and Yong Zhao
- Subjects
0301 basic medicine ,Small interfering RNA ,AMP-Activated Protein Kinases ,Neuroprotection ,Article ,Brain Ischemia ,03 medical and health sciences ,AMP-activated protein kinase ,medicine ,Animals ,Humans ,Gene Silencing ,RNA, Small Interfering ,Organelle Biogenesis ,Multidisciplinary ,biology ,Chemistry ,Nuclear Proteins ,AMPK ,Infarction, Middle Cerebral Artery ,TFAM ,medicine.disease ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Mitochondria ,Rats ,Cell biology ,Stroke ,Disease Models, Animal ,030104 developmental biology ,Mitochondrial biogenesis ,Reperfusion Injury ,biology.protein ,Organelle biogenesis ,Reperfusion injury ,Signal Transduction - Abstract
Sestrin2 (Sesn2) exerts neuroprotective properties in some neurodegenerative diseases. However, the role of Sesn2 in stroke is unclear. The AMP-activated protein kinase/peroxisome proliferator-activated receptor γ coactivator-1α (AMPK/PGC-1α) pathway plays an important role in regulating mitochondrial biogenesis, which helps prevent cerebral ischemia/reperfusion (I/R) injury. Here, we aimed to determine whether Sesn2 alleviated I/R damage by regulating mitochondrial biogenesis through the AMPK/PGC-1α signaling pathway. To be able to test this, Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 1 h with Sesn2 silencing. At 24 h after reperfusion, we found that neurological deficits were exacerbated, infarct volume was enlarged, and oxidative stress and neuronal damage were greater in the Sesn2 siRNA group than in the MCAO group. To explore protective mechanisms, an AMPK activator was used. Expression levels of Sesn2, p-AMPK, PGC-1α, NRF-1, TFAM, SOD2, and UCP2 were significantly increased following cerebral I/R. However, upregulation of these proteins was prevented by Sesn2 small interfering RNA (siRNA). In contrast, activation of AMPK with 5′-aminoimidazole-4-carboxamide riboside weakened the effects of Sesn2 siRNA. These results suggest that Sesn2 silencing may suppress mitochondrial biogenesis, reduce mitochondrial biological activity, and finally aggravate cerebral I/R injury through inhibiting the AMPK/PGC-1α pathway.
- Published
- 2016
12. Foxo1-mediated inflammatory response after cerebral hemorrhage in rats
- Author
-
Zhenyu Li, Qi He, Yan You, Yanghao Hou, Yong Zhao, Jing Zhao, Xuan Zhai, Faming He, and Lingyu Li
- Subjects
0301 basic medicine ,Male ,endocrine system ,Interleukin-1beta ,FOXO1 ,Nerve Tissue Proteins ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Medicine ,Animals ,Cerebral Hemorrhage ,Peroxidase ,Intracerebral hemorrhage ,Gene knockdown ,business.industry ,General Neuroscience ,Interleukin-18 ,nutritional and metabolic diseases ,food and beverages ,FOXO Family ,Brain ,NF-κB ,medicine.disease ,Rats ,030104 developmental biology ,chemistry ,Apoptosis ,Immunology ,Cancer research ,TLR4 ,Encephalitis ,lipids (amino acids, peptides, and proteins) ,Interleukin 18 ,business ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery - Abstract
The forkhead box O (Foxo) family of transcription factors plays a crucial role in cell apoptosis, immune regulation, and tissue development. Foxo1, as the foremost member of the Foxo family, regulates a wide range of molecular signals in many tissues, including tumor, liver, and brain. This study investigated Foxo1 expression at different time points and in different brain areas, and the role of Foxo1 in vivo in regulating inflammatory injury in a rat model of autologous blood-injected cerebral hemorrhage injury. We found that Foxo1 expression peaked at 12h post-intracerebral hemorrhage (ICH) and in the ipsilateral corpus striatum. Foxo1 knockdown by Foxo1 siRNA decreased ICH injury, improved neurological function, and decreased the expression of inflammatory factors downstream of the Foxo1 pathway, including TLR4, NF-κB, TNF-α, IL-1β, and IL-18. Foxo1 knockdown also decreased the expression and activity of myeloperoxidase, IL-1β, and IL-18. In conclusion, our findings demonstrate that Foxo1 is a key regulator of inflammatory injury in rats after ICH. By identifying the molecular mechanisms of Foxo1/TLR4/NF-κB signaling, we provide a novel rationale for therapeutic approaches to managing inflammatory injury after ICH.
- Published
- 2016
13. Protective effects of PGC-1α via the mitochondrial pathway in rat brains after intracerebral hemorrhage
- Author
-
Yong Zhao, Xuan Zhai, Zhenyu Li, Yanghao Hou, Yan You, Lingyu Li, and Jing Zhao
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,SOD2 ,Pharmacology ,Mitochondrion ,Biology ,medicine.disease_cause ,Neuroprotection ,DNA, Mitochondrial ,Mitochondrial Proteins ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Adenosine Triphosphate ,medicine ,Animals ,cardiovascular diseases ,NRF1 ,RNA, Messenger ,Molecular Biology ,Cerebral Hemorrhage ,Intracerebral hemorrhage ,General Neuroscience ,Brain ,TFAM ,medicine.disease ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,nervous system diseases ,Mitochondria ,Rats ,030104 developmental biology ,Mitochondrial biogenesis ,Neurology (clinical) ,030217 neurology & neurosurgery ,Oxidative stress ,Developmental Biology - Abstract
Peroxisome-proliferator-activated receptor co-activator-1α (PGC-1α) is a transcriptional co-activator that coordinately regulates genes required for mitochondrial biogenesis, which stimulates mitochondrial activity. It is also a major factor in the up-regulation of antioxidant activities that are a response to oxidative stress. However, the role of PGC-1α after intracerebral hemorrhage (ICH) has not been studied. The purpose of the present work was to investigate the effects and mechanism of PGC-1α after ICH in the brain. Brain injury was induced by injecting autologous arterial blood (50 μL) into the rat brain. PGC-1α siRNAs were injected into rat brains 24 h prior to ICH. Then, 72 h after ICH, brains were collected for investigation. Post-assessment included western blot analysis, RT-PCR assay, neurobehavioral function testing, measurement of brain water content, high-performance liquid chromatography (HPLC), and projection electron microscopy on ICH rat models. The concentration of PGC-1α was higher in the ipsilateral striatum after ICH, peaking around 72 h after ICH. The expression of NRF-1, TFAM, SOD2, UCP2, mitochondrial DNA, ATP concentration, mitochondrial quantity, and brain water content were increased 72 h after ICH. However, the neurological score was decreased 72 h after ICH. Treatment with PGC-1α siRNAs significantly decreased the neurological score, ATP concentration, number of mitochondria, expression of NRF-1, TFAM, SOD2, UCP2, and mitochondrial DNA, and increased brain water content and formation of mitochondrial myelin layer structures. In conclusion, our data suggest that PGC-1α protects rat brains via a mitochondrial pathway following ICH. Key words: PGC-1α intracerebral hemorrhage(ICH); mitochondrial; neuroprotection.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.