1
|
Fu G, Kang X, Lin S. Glycyrrhizic Acid Inhibits Hippocampal Neuron Apoptosis by Activating the PI3K/ AKT Signaling Pathway. Biochem Genet 2024:10.1007/s10528-024-10936-w. [PMID: 39377899 DOI: 10.1007/s10528-024-10936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024]
Abstract
Glycyrrhizic acid (GA), one of the main active substances in Glycyrrhiza, has anti-inflammatory, anti-viral, and neuroprotective effects. GA can significantly reduce cerebral infarction size in middle cerebral artery occlusion (MCAO) rats and suppress inflammatory responses. However, the underlying mechanism by which GA protects the neuronal system remains poorly understood. Cell proliferation and viability were tested using CCK-8 and Edu assays. The effects of GA on apoptosis were detected using flow cytometry and Tunel assays. Western blotting was performed to assess protein expression. Behavioral experiments were conducted using the Morris water maze and rotation tests. Infarct size was observed using TTC staining. We report that GA protects neurons by inhibiting apoptosis, mainly through the PI3K/AKT pathway in oxygen-glucose deprivation/reoxygenation (OGD/R) and MCAO rat models. GA increases the viability and proliferation of oxygen- and glucose-deprived hippocampal neurons. Hippocampal neuron apoptosis decreased after GA treatment in vitro and in vivo. Furthermore, we determined that GA treatment increased the active state of PI3K and its downstream protein p-AKT, whereas when using a specific inhibitor of PI3K, Y294002, the levels of p-PI3K and p-AKT decreased. Finally, we showed that GA treatment improved spatial memory and motor coordination in MCAO rats, while TTC staining showed that GA decreased cerebral infarct size in MCAO rats. We reveal that GA protects hippocampal neurons by inhibiting their apoptosis, mainly through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Guanglei Fu
- Infectious Disease Department, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xuedi Kang
- The Fourth Clinical Medical College & ShenZhen Traditional Chinese Medicine Hospital, GuangZhou University of Chinese Medicine, 1 Fuhua Road, Futian District, Shenzhen, China.
| | - Songjun Lin
- The Fourth Clinical Medical College & ShenZhen Traditional Chinese Medicine Hospital, GuangZhou University of Chinese Medicine, 1 Fuhua Road, Futian District, Shenzhen, China.
- Encephalopathy and Psychology Department, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical MedicalCollege of Guangzhou University of Chinese Medicine, 1 Fuhua Road, Futian District, Shenzhen, 518000, China.
| |
Collapse
|
2
|
Boisserand LSB, Geraldo LH, Bouchart J, El Kamouh MR, Lee S, Sanganahalli BG, Spajer M, Zhang S, Lee S, Parent M, Xue Y, Skarica M, Yin X, Guegan J, Boyé K, Saceanu Leser F, Jacob L, Poulet M, Li M, Liu X, Velazquez SE, Singhabahu R, Robinson ME, Askenase MH, Osherov A, Sestan N, Zhou J, Alitalo K, Song E, Eichmann A, Sansing LH, Benveniste H, Hyder F, Thomas JL. VEGF-C prophylaxis favors lymphatic drainage and modulates neuroinflammation in a stroke model. J Exp Med 2024; 221:e20221983. [PMID: 38442272 PMCID: PMC10913814 DOI: 10.1084/jem.20221983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 11/13/2023] [Accepted: 01/25/2024] [Indexed: 03/07/2024] Open
Abstract
Meningeal lymphatic vessels (MLVs) promote tissue clearance and immune surveillance in the central nervous system (CNS). Vascular endothelial growth factor-C (VEGF-C) regulates MLV development and maintenance and has therapeutic potential for treating neurological disorders. Herein, we investigated the effects of VEGF-C overexpression on brain fluid drainage and ischemic stroke outcomes in mice. Intracerebrospinal administration of an adeno-associated virus expressing mouse full-length VEGF-C (AAV-mVEGF-C) increased CSF drainage to the deep cervical lymph nodes (dCLNs) by enhancing lymphatic growth and upregulated neuroprotective signaling pathways identified by single nuclei RNA sequencing of brain cells. In a mouse model of ischemic stroke, AAV-mVEGF-C pretreatment reduced stroke injury and ameliorated motor performances in the subacute stage, associated with mitigated microglia-mediated inflammation and increased BDNF signaling in brain cells. Neuroprotective effects of VEGF-C were lost upon cauterization of the dCLN afferent lymphatics and not mimicked by acute post-stroke VEGF-C injection. We conclude that VEGF-C prophylaxis promotes multiple vascular, immune, and neural responses that culminate in a protection against neurological damage in acute ischemic stroke.
Collapse
Affiliation(s)
| | - Luiz Henrique Geraldo
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Jean Bouchart
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Marie-Renee El Kamouh
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France
| | - Seyoung Lee
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Myriam Spajer
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France
| | - Shenqi Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Sungwoon Lee
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Maxime Parent
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Yuechuan Xue
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Mario Skarica
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Xiangyun Yin
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Justine Guegan
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France
| | - Kevin Boyé
- Paris Cardiovascular Research Center, INSERM U970, Paris, France
| | - Felipe Saceanu Leser
- Paris Cardiovascular Research Center, INSERM U970, Paris, France
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laurent Jacob
- Paris Cardiovascular Research Center, INSERM U970, Paris, France
| | - Mathilde Poulet
- Paris Cardiovascular Research Center, INSERM U970, Paris, France
| | - Mingfeng Li
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Xiodan Liu
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Sofia E. Velazquez
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Ruchith Singhabahu
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France
| | - Mark E. Robinson
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | | | - Artem Osherov
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT, USA
| | - Jiangbing Zhou
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Kari Alitalo
- Faculty of Medicine, Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Eric Song
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
- Paris Cardiovascular Research Center, INSERM U970, Paris, France
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris, France
| |
Collapse
|
3
|
Lian Z, Yu SR, Cui YX, Li SF, Su L, Song JX, Lee CY, Chen QX, Chen H. Rosuvastatin Enhances Lymphangiogenesis after Myocardial Infarction by Regulating the miRNAs/Vascular Endothelial Growth Factor Receptor 3 (miRNAs/VEGFR3) Pathway. ACS Pharmacol Transl Sci 2024; 7:335-347. [PMID: 38357274 PMCID: PMC10863446 DOI: 10.1021/acsptsci.3c00151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Several clinical studies have suggested that the early administration of statins could reduce the risk of in-hospital mortality in acute myocardial infarction (AMI) patients. Recently, some studies have identified that stimulating lymphangiogenesis after AMI could improve cardiac function by reducing myocardial edema and inflammation. This study aimed to identify the effect of rosuvastatin on postinfarct lymphangiogenesis and to identify the underlying mechanism of this effect. METHOD Myocardial infarction (MI) was induced by ligation of the left anterior descending coronary artery in mice orally administered rosuvastatin for 7 days. The changes in cardiac function, pathology, and lymphangiogenesis following MI were measured by echocardiography and immunostaining. EdU, Matrigel tube formation, and scratch wound assays were used to evaluate the effect of rosuvastatin on the proliferation, tube formation, and migration of the lymphatic endothelial cell line SVEC4-10. The expression of miR-107-3p, miR-491-5p, and VEGFR3 was measured by polymerase chain reaction (PCR) and Western blotting. A gain-of-function study was performed using miR-107-3p and miR-491-5p mimics. RESULTS The rosuvastatin-treated mice had a significantly improved ejection fraction and increased lymphatic plexus density 7 days after MI. Rosuvastatin also reduced myocardial edema and inflammatory response after MI. We used a VEGFR3 inhibitor to partially reverse these effects. Rosuvastatin promoted the proliferation, migration, and tube formation of SVEC4-10 cells. PCR and Western blot analyses revealed that rosuvastatin intervention downregulated miR-107-3p and miR-491-5p and promoted VEGFR3 expression. The gain-of-function study showed that miR-107-3p and miR-491-5p could inhibit the proliferation, migration, and tube formation of SVEC4-10 cells. CONCLUSION Rosuvastatin could improve heart function by promoting lymphangiogenesis after MI by regulating the miRNAs/VEGFR3 pathway.
Collapse
Affiliation(s)
- Zheng Lian
- Cardiovascular
Center, Beijing Tongren Hospital, Capital
Medical University, Xihuan South Road No. 2, Economic-Technological
Development Area, Beijing 100176, China
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Shi-Ran Yu
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Yu-Xia Cui
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Su-Fang Li
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Li−Na Su
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Jun-Xian Song
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Chong-Yoo Lee
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Qi-Xin Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Hong Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| |
Collapse
|
4
|
Simoes Braga Boisserand L, Bouchart J, Geraldo LH, Lee S, Sanganahalli BG, Parent M, Zhang S, Xue Y, Skarica M, Guegan J, Li M, Liu X, Poulet M, Askanase M, Osherov A, Spajer M, Kamouh MRE, Eichmann A, Alitalo K, Zhou J, Sestan N, Sansing LH, Benveniste H, Hyder F, Thomas JL. VEGF-C promotes brain-derived fluid drainage, confers neuroprotection, and improves stroke outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542708. [PMID: 37398128 PMCID: PMC10312491 DOI: 10.1101/2023.05.30.542708] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Meningeal lymphatic vessels promote tissue clearance and immune surveillance in the central nervous system (CNS). Vascular endothelium growth factor-C (VEGF-C) is essential for meningeal lymphatic development and maintenance and has therapeutic potential for treating neurological disorders, including ischemic stroke. We have investigated the effects of VEGF-C overexpression on brain fluid drainage, single cell transcriptome in the brain, and stroke outcomes in adult mice. Intra-cerebrospinal fluid administration of an adeno-associated virus expressing VEGF-C (AAV-VEGF-C) increases the CNS lymphatic network. Post-contrast T1 mapping of the head and neck showed that deep cervical lymph node size and drainage of CNS-derived fluids were increased. Single nuclei RNA sequencing revealed a neuro-supportive role of VEGF-C via upregulation of calcium and brain-derived neurotrophic factor (BDNF) signaling pathways in brain cells. In a mouse model of ischemic stroke, AAV-VEGF-C pretreatment reduced stroke injury and ameliorated motor performances in the subacute stage. AAV-VEGF-C thus promotes CNS-derived fluid and solute drainage, confers neuroprotection, and reduces ischemic stroke damage. Short abstract Intrathecal delivery of VEGF-C increases the lymphatic drainage of brain-derived fluids confers neuroprotection, and improves neurological outcomes after ischemic stroke.
Collapse
|
5
|
Sousa CS, Lima R, Cibrão JR, Gomes ED, Fernandes LS, Pinho TS, Silva D, Campos J, Salgado AJ, Silva NA. Pre-Clinical Assessment of Roflumilast Therapy in a Thoracic Model of Spinal Cord Injury. Pharmaceutics 2023; 15:1556. [PMID: 37242797 PMCID: PMC10222626 DOI: 10.3390/pharmaceutics15051556] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
The failure of axons to regenerate after a spinal cord injury (SCI) remains one of the greatest challenges in neuroscience. The initial mechanical trauma is followed by a secondary injury cascade, creating a hostile microenvironment, which not only is not permissive to regeneration but also leads to further damage. One of the most promising approaches for promoting axonal regeneration is to maintain the levels of cyclic adenosine monophosphate (cAMP), specifically by a phosphodiesterase-4 (PDE4) inhibitor expressed in neural tissues. Therefore, in our study, we evaluated the therapeutic effect of an FDA-approved PDE4 inhibitor, Roflumilast (Rof), in a thoracic contusion rat model. Results indicate that the treatment was effective in promoting functional recovery. Rof-treated animals showed improvements in both gross and fine motor function. Eight weeks post-injury, the animals significantly recovered by achieving occasional weight-supported plantar steps. Histological assessment revealed a significant decrease in cavity size, less reactive microglia, as well as higher axonal regeneration in treated animals. Molecular analysis revealed that IL-10 and IL-13 levels, as well as VEGF, were increased in the serum of Rof-treated animals. Overall, Roflumilast promotes functional recovery and supports neuroregeneration in a severe thoracic contusion injury model and may be important in SCI treatment.
Collapse
Affiliation(s)
- Carla S Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
- Department of Neurosurgery, Hospital Garcia de Orta, 2805-267 Almada, Portugal
| | - Rui Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Jorge R Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Luís S Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Deolinda Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| |
Collapse
|
6
|
The Lymphatic Endothelium in the Context of Radioimmuno-Oncology. Cancers (Basel) 2022; 15:cancers15010021. [PMID: 36612017 PMCID: PMC9817924 DOI: 10.3390/cancers15010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The study of lymphatic tumor vasculature has been gaining interest in the context of cancer immunotherapy. These vessels constitute conduits for immune cells' transit toward the lymph nodes, and they endow tumors with routes to metastasize to the lymph nodes and, from them, toward distant sites. In addition, this vasculature participates in the modulation of the immune response directly through the interaction with tumor-infiltrating leukocytes and indirectly through the secretion of cytokines and chemokines that attract leukocytes and tumor cells. Radiotherapy constitutes the therapeutic option for more than 50% of solid tumors. Besides impacting transformed cells, RT affects stromal cells such as endothelial and immune cells. Mature lymphatic endothelial cells are resistant to RT, but we do not know to what extent RT may affect tumor-aberrant lymphatics. RT compromises lymphatic integrity and functionality, and it is a risk factor to the onset of lymphedema, a condition characterized by deficient lymphatic drainage and compromised tissue homeostasis. This review aims to provide evidence of RT's effects on tumor vessels, particularly on lymphatic endothelial cell physiology and immune properties. We will also explore the therapeutic options available so far to modulate signaling through lymphatic endothelial cell receptors and their repercussions on tumor immune cells in the context of cancer. There is a need for careful consideration of the RT dosage to come to terms with the participation of the lymphatic vasculature in anti-tumor response. Here, we provide new approaches to enhance the contribution of the lymphatic endothelium to radioimmuno-oncology.
Collapse
|
7
|
Scott XO, Chen SH, Hadad R, Yavagal D, Peterson EC, Starke RM, Dietrich WD, Keane RW, de Rivero Vaccari JP. Cohort study on the differential expression of inflammatory and angiogenic factors in thrombi, cerebral and peripheral plasma following acute large vessel occlusion stroke. J Cereb Blood Flow Metab 2022; 42:1827-1839. [PMID: 35673992 PMCID: PMC9536118 DOI: 10.1177/0271678x221106956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022]
Abstract
Inflammation plays an important role in the pathogenesis of stroke. The differential expression of inflammatory and angiogenic factors in thrombi and plasma remain undefined. In this observational cohort study, we evaluated angiogenic factors and inflammatory cytokines, in cerebral thrombi, local cerebral plasma (CP), and peripheral plasma (PP) in patients with acute ischemic stroke. Protein analysis of thrombi, CP and PP were used to measure angiogenic and inflammatory proteins using electrochemiluminescence. Our data indicate that VEGF-A, VEGF-C, bFGF, IL-4, IL-13, IL-1β, IL-2, IL-8, IL-16, IL-6 and IL-12p70 were higher in the thrombi of acute ischemic stroke patients than in the CP and PP of stroke patients. Moreover, the protein levels of GM-CSF were lower in the PP than in the CP and the clot. Moreover, VEGF-D, Flt-1, PIGF, TIE-2, IL-5, TNF-β, IL-15, IL-12/IL-23p40, IFN-γ and IL-17A were higher in PP and CP than in thrombi. Our results show that cytokines mediating the inflammatory response and proteins involved in angiogenesis are differentially expressed in thrombi within the cerebral and peripheral circulations. These data highlight the importance of identifying new biomarkers in different compartments of the circulatory system and in thrombi that may be used for the diagnosis and treatment of stroke patients.
Collapse
Affiliation(s)
- Xavier O Scott
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephanie H Chen
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dileep Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eric C Peterson
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert M Starke
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert W Keane
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
8
|
Ganesana M, Venton BJ. Spontaneous, transient adenosine release is not enhanced in the CA1 region of hippocampus during severe ischemia models. J Neurochem 2021; 159:887-900. [PMID: 34453336 PMCID: PMC8627433 DOI: 10.1111/jnc.15496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/23/2022]
Abstract
Ischemic stroke causes damage in the brain, and a slow buildup of adenosine is neuroprotective during ischemic injury. Spontaneous, transient adenosine signaling, lasting only 3 s per event, has been discovered that increases in frequency in the caudate-putamen during early stages of mild ischemia-reperfusion injury. However, spontaneous adenosine changes have not been studied in the hippocampus during ischemia, an area highly susceptible to stroke. Here, we investigated changes of spontaneous, transient adenosine in the CA1 region of rat hippocampus during three different models of the varied intensity of ischemia. During the early stages of the milder bilateral common carotid artery occlusion (BCCAO) model, there were fewer spontaneous, transient adenosine, but no change in the concentration of individual events. In contrast, during the moderate 2 vertebral artery occlusion (2VAO) and severe 4 vessel occlusion (4VO) models, both the frequency of spontaneous, transient adenosine and the average event adenosine concentration decreased. Blood flow measurements validate that the ischemia models decreased blood flow, and corresponding pathological changes were observed by transmission electron microscopy (TEM). 4VO occlusion showed the most severe damage in histology and BCCAO showed the least. Overall, our data suggest that there is no enhanced spontaneous adenosine release in the hippocampus during moderate and severe ischemia, which could be due to depletion of the rapidly releasable adenosine pool. Thus, during ischemic stroke, there are fewer spontaneous adenosine events that could inhibit neurotransmission, which might lead to more damage and less neuroprotection in the hippocampus CA1 region. Read the Editorial Highlight for this article on page 800.
Collapse
Affiliation(s)
- Mallikarjunarao Ganesana
- Department of Chemistry and Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
| | - B. Jill Venton
- Department of Chemistry and Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
9
|
Jeong KH, Cho KO, Lee MY, Kim SY, Kim WJ. Vascular endothelial growth factor receptor-3 regulates astroglial glutamate transporter-1 expression via mTOR activation in reactive astrocytes following pilocarpine-induced status epilepticus. Glia 2020; 69:296-309. [PMID: 32835451 DOI: 10.1002/glia.23897] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022]
Abstract
Recent evidence has shown that the vascular endothelial growth factor (VEGF) system plays a crucial role in several neuropathological processes. We previously reported an upregulation of VEGF-C and its receptor, VEGFR-3, in reactive astrocytes after the onset of status epilepticus (SE). However, it remains unknown, which molecules act as downstream signals following VEGFR-3 upregulation, and are involved in reactive astrogliosis after SE. Therefore, we investigated whether VEGFR-3 upregulation within reactive astrocytes is associated with the activation of mammalian target of rapamycin (mTOR) signaling, which we confirmed by assaying for the phosphorylated form of S6 protein (pS6), and whether VEGFR-3-mediated mTOR activation induces astroglial glutamate transporter-1 (GLT-1) expression in the hippocampus after pilocarpine-induced SE. We found that spatiotemporal expression of pS6 was consistent with VEGFR-3 expression in the hippocampus after SE, and that both pS6 and VEGFR-3 were highly expressed in SE-induced reactive astrocytes. Treatment with the mTOR inhibitor rapamycin decreased astroglial VEGFR-3 expression and GLT-1 expression after SE. Treatment with a selective inhibitor for VEGFR-3 attenuated astroglial pS6 expression as well as suppressed GLT-1 expression and astroglial reactivity in the hippocampus after SE. These findings demonstrate that VEGFR-3-mediated mTOR activation could contribute to the regulation of GLT-1 expression in reactive astrocytes during the subacute phase of epilepsy. In conclusion, the present study suggests that VEGFR-3 upregulation in reactive astrocytes may play a role in preventing hyperexcitability induced by continued seizure activity.
Collapse
Affiliation(s)
- Kyoung Hoon Jeong
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyung-Ok Cho
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mun-Yong Lee
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong Yun Kim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Won-Joo Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Phthalide derivative CD21 ameliorates ischemic brain injury in a mouse model of global cerebral ischemia: involvement of inhibition of NLRP3. Int Immunopharmacol 2020; 86:106714. [PMID: 32593156 DOI: 10.1016/j.intimp.2020.106714] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/28/2022]
Abstract
The activation of NLRP3 inflammasome is closely related to ischemic brain injury and inhibition of NLRP3 inflammasome activation may be a new therapeutic strategy for ischemic stroke. Our previous studies showed that ligustilide (LIG) had a dose-dependent neuroprotective effect on various models of cerebral ischemia and dementia in vivo and in vitro. CD21, a kind of phthalide derivative, was modified from LIG. In this study, we established a global cerebral ischemia-reperfusion model in mice by bilateral common carotid artery ligation (2VO), and explored the neuroprotective effect of CD21 and its anti-inflammatory mechanism on cerebral ischemia mice. CD21 significantly improved weight loss, neurobehavioral deficits and neurons loss in hippocampal CA1 and caudate putamen (CPu) subregions, which were induced by 2VO in mice. CD21 significantly inhibited the overactivation of astrocyte and microglia, and decreased the mRNA level of IL-6, TNF-α and IL-1β. Moreover, CD21 significantly inhibited the activation of TLR4/NF-κB signaling pathway mediated by HMGB1 and NLRP3/ASC/Caspase-1 signaling pathway mediated by Cathepsin B, thus inhibiting the activation of NLRP3 inflammasome. Our results demonstrated that CD21 may exert a neuroprotection by inhibiting NLRP3 inflammasome activation after cerebral ischemia. These findings provide a new strategy for the treatment of ischemic stroke.
Collapse
|
11
|
Pinho AG, Cibrão JR, Silva NA, Monteiro S, Salgado AJ. Cell Secretome: Basic Insights and Therapeutic Opportunities for CNS Disorders. Pharmaceuticals (Basel) 2020; 13:E31. [PMID: 32093352 PMCID: PMC7169381 DOI: 10.3390/ph13020031] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
Transplantation of stem cells, in particular mesenchymal stem cells (MSCs), stands as a promising therapy for trauma, stroke or neurodegenerative conditions such as spinal cord or traumatic brain injuries (SCI or TBI), ischemic stroke (IS), or Parkinson's disease (PD). Over the last few years, cell transplantation-based approaches have started to focus on the use of cell byproducts, with a strong emphasis on cell secretome. Having this in mind, the present review discusses the current state of the art of secretome-based therapy applications in different central nervous system (CNS) pathologies. For this purpose, the following topics are discussed: (1) What are the main cell secretome sources, composition, and associated collection techniques; (2) Possible differences of the therapeutic potential of the protein and vesicular fraction of the secretome; and (3) Impact of the cell secretome on CNS-related problems such as SCI, TBI, IS, and PD. With this, we aim to clarify some of the main questions that currently exist in the field of secretome-based therapies and consequently gain new knowledge that may help in the clinical application of secretome in CNS disorders.
Collapse
Affiliation(s)
- Andreia G. Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Jorge R. Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nuno A. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| |
Collapse
|
12
|
Yanev P, Poinsatte K, Hominick D, Khurana N, Zuurbier KR, Berndt M, Plautz EJ, Dellinger MT, Stowe AM. Impaired meningeal lymphatic vessel development worsens stroke outcome. J Cereb Blood Flow Metab 2020; 40:263-275. [PMID: 30621519 PMCID: PMC7370617 DOI: 10.1177/0271678x18822921] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The discovery of meningeal lymphatic vessels (LVs) has sparked interest in identifying their role in diseases of the central nervous system. Similar to peripheral LVs, meningeal LVs depend on vascular endothelial growth factor receptor-3 (VEGFR3) signaling for development. Here we characterize the effect of stroke on meningeal LVs, and the impact of meningeal lymphatic hypoplasia on post-stroke outcomes. We show that photothrombosis (PT), but not transient middle cerebral artery occlusion (tMCAo), induces meningeal lymphangiogenesis in young male C57Bl/J6 mice. We also show that Vegfr3wt/mut mice develop significantly fewer meningeal LVs than Vegfr3wt/wt mice. Again, meningeal lymphangiogenesis occurs in the alymphatic zone lateral to the sagittal sinus only after PT-induced stroke in Vegfr3wt/wt mice. Interestingly, Vegfr3wt/mut mice develop larger stroke volumes than Vegfr3wt/wt mice after tMCAo, but not after PT. Our results reveal differences between PT and tMCAo models of stroke and underscore the need to consider method of stroke induction when investigating the role of meningeal lymphatics. Taken together, our data indicate that ischemic injury can induce the growth of meningeal LVs and that the absence of these LVs can impact post-stroke outcomes.
Collapse
Affiliation(s)
- Pavel Yanev
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Katherine Poinsatte
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Devon Hominick
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noor Khurana
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kielen R Zuurbier
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcus Berndt
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Erik J Plautz
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael T Dellinger
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Neurology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
13
|
Cho KO, Kim JY, Jeong KH, Lee MY, Kim SY. Increased expression of vascular endothelial growth factor-C and vascular endothelial growth factor receptor-3 after pilocarpine-induced status epilepticus in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:281-289. [PMID: 31297012 PMCID: PMC6609264 DOI: 10.4196/kjpp.2019.23.4.281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
Abstract
Vascular endothelial growth factor (VEGF)-C and its receptor, vascular endothelial growth factor receptor (VEGFR)-3, are responsible for lymphangiogenesis in both embryos and adults. In epilepsy, the expression of VEGF-C and VEGFR-3 was significantly upregulated in the human brains affected with temporal lobe epilepsy. Moreover, pharmacologic inhibition of VEGF receptors after acute seizures could suppress the generation of spontaneous recurrent seizures, suggesting a critical role of VEGF-related signaling in epilepsy. Therefore, in the present study, the spatiotemporal expression of VEGF-C and VEGFR-3 against pilocarpine-induced status epilepticus (SE) was investigated in C57BL/6N mice using immunohistochemistry. At 1 day after SE, hippocampal astrocytes and microglia were activated. Pyramidal neuronal death was observed at 4 days after SE. In the subpyramidal zone, VEGF-C expression gradually increased and peaked at 7 days after SE, while VEGFR-3 was significantly upregulated at 4 days after SE and began to decrease at 7 days after SE. Most VEGF-C/VEGFR-3-expressing cells were pyramidal neurons, but VEGF-C was also observed in some astrocytes in sham-manipulated animals. However, at 4 days and 7 days after SE, both VEGFR-3 and VEGF-C immunoreactivities were observed mainly in astrocytes and in some microglia of the stratum radiatum and lacunosum-moleculare of the hippocampus, respectively. These data indicate that VEGF-C and VEGFR-3 can be upregulated in hippocampal astrocytes and microglia after pilocarpine-induced SE, providing basic information about VEGF-C and VEGFR-3 expression patterns following acute seizures.
Collapse
Affiliation(s)
- Kyung-Ok Cho
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Institute of Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Joo Youn Kim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Kyoung Hoon Jeong
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Mun-Yong Lee
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seong Yun Kim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
14
|
Wen MD, Jiang Y, Huang J, Al-Hawwas M, Dan QQ, Yang RA, Yuan B, Zhao XM, Jiang L, Zhong MM, Xiong LL, Zhang YH. A Novel Role of VEGFC in Cerebral Ischemia With Lung Injury. Front Neurosci 2019; 13:479. [PMID: 31191213 PMCID: PMC6540825 DOI: 10.3389/fnins.2019.00479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 04/26/2019] [Indexed: 02/05/2023] Open
Abstract
Cerebral ischemia (CI) is a severe brain injury resulting in a variety of motor impairments combined with secondary injury in remote organs, especially the lung. This condition occurs due to insufficient blood supply to the brain during infancy. However, it has a molecular linkage that needs to be thoroughly covered. Here, we report on the role of vascular endothelial growth factor C (VEGFC) in lung injury induced by CI. The middle cerebral artery occlusion (MCAO) was depended to establish the animal model of CI. Rats were used and brain ischemia was confirmed through TTC staining. Serum was used for protein chip analysis to study the proteomic interaction. Immunohistochemistry analyses were used to quantify and locate the VEGFC in the lung and brain. The role of VEGFC was detected by siVEGFC technology in SY5Y, HUCEV, and A549 cell lines, under normal and oxygen glucose deprivation (OGD) conditions in vitro. As a result, the TTC staining demonstrated that the model of brain ischemia was successfully established, and MPO experiments reported that lung damage was induced in MCAO rats. VEGFC levels were up-regulated in serum. On the other hand, immunohistochemistry showed that VEGFC increased significantly in the cytoplasm of neurons, the endothelium of small trachea and the lung cells of CI animals. On a functional level, siVEGFC effectively inhibited the proliferation of SY5Y cells and decreased the viability of HUVEC cells in normal cell lines. But under OGD conditions, siVEGFC decreased the growth of HUVEC and increased the viability of A549 cells, while no effect was noticed on SYSY cells. Therefore, we confirmed the different role of VEGFC played in neurons and lung cells in cerebral ischemia-reperfusion injury. These findings may contribute to the understanding the molecular linkage of brain ischemia and lung injury, which therefore provides a new idea for the therapeutic approach to cerebral ischemia-reperfusion.
Collapse
Affiliation(s)
- Mu-Dong Wen
- Department of Respiration, The First People's Hospital of Yunnan Province, Kunming, China
| | - Ya Jiang
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Jin Huang
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Mohammed Al-Hawwas
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Qi-Qin Dan
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rui-An Yang
- Department of Respiration, The First People's Hospital of Yunnan Province, Kunming, China
| | - Bing Yuan
- Department of Respiration, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xiao-Ming Zhao
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Jiang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ming-Mei Zhong
- Department of Respiration, The First People's Hospital of Yunnan Province, Kunming, China
| | - Liu-Lin Xiong
- Department of Anesthesiology, National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Yun-Hui Zhang
- Department of Respiration, The First People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
15
|
Wahul AB, Joshi PC, Kumar A, Chakravarty S. Transient global cerebral ischemia differentially affects cortex, striatum and hippocampus in Bilateral Common Carotid Arterial occlusion (BCCAo) mouse model. J Chem Neuroanat 2018; 92:1-15. [DOI: 10.1016/j.jchemneu.2018.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/24/2022]
|
16
|
Zhou XB, Zou DX, Gu W, Wang D, Feng JS, Wang JY, Zhou JL. An Experimental Study on Repeated Brief Ischemia in Promoting Sciatic Nerve Repair and Regeneration in Rats. World Neurosurg 2018; 114:e11-e21. [PMID: 29374605 DOI: 10.1016/j.wneu.2018.01.125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Research has shown that ischemic preconditioning reduced the severity of ischemia-reperfusion injury in brain in rats, we have a hypothesis that repeated brief ischemia has positive effects on peripheral nerve damage. This study was conducted to investigate the potential protective effects of repeated brief ischemia on peripheral nerve regeneration using a rat model of experimental sciatic nerve transection injury. METHODS Treatment groups (groups A-D) received repeated, brief ischemia every 1 day/2 days/3 days/7 days. After surgery for 4, 8, 12 weeks, we evaluated sciatic functional index test, gastrocnemius muscle wet mass, axon and nerve fiber diameter, density, G-ratio, immunohistochemistry of S-100, vascular endothelial growth factor (VEGF), and the ultrastructure of the nerves. RESULTS Sciatic functional index test and muscle wet mass were improved on the repeated brief ischemia groups. Ischemia treatment resulted in a significant increase in axon and nerve fiber density as well as S-100 and VEGF-positive cell, which indicated that repeated brief ischemia promotes Schwann cell proliferation and reconstruction. CONCLUSIONS This study exhibits the positive effects of repeated brief ischemia in sciatic nerve transection injury, possibly in part because it can improve VEGF and the physiologic state of Schwann cells in the ischemic environment and then accelerate the ability of neurite outgrow.
Collapse
Affiliation(s)
- Xiao-Bin Zhou
- Department of Orthopedics, The Third Hospital of Shi Jia-Zhuang, Hebei, People's Republic of China; Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - De-Xin Zou
- Department of Spine Surgery, YanTai-Shan Hospital, Shandong, People's Republic of China
| | - Wei Gu
- Department of Ophthalmology, The Third Hospital of Shi Jia-Zhuang, Hebei, People's Republic of China
| | - Dong Wang
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jian-Shu Feng
- Department of Orthopedics, The Third Hospital of Shi Jia-Zhuang, Hebei, People's Republic of China
| | - Jiang-Yong Wang
- Department of Orthopedics, The Third Hospital of Shi Jia-Zhuang, Hebei, People's Republic of China
| | - Jun-Lin Zhou
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
17
|
Zhou HJ, Li H, Shi MQ, Mao XN, Liu DL, Chang YR, Gan YM, Kuang X, Du JR. Protective Effect of Klotho against Ischemic Brain Injury Is Associated with Inhibition of RIG-I/NF-κB Signaling. Front Pharmacol 2018; 8:950. [PMID: 29403373 PMCID: PMC5778393 DOI: 10.3389/fphar.2017.00950] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/14/2017] [Indexed: 01/19/2023] Open
Abstract
Aging is the greatest independent risk factor for the occurrence of stroke and poor outcomes, at least partially through progressive increases in oxidative stress and inflammation with advanced age. Klotho is an antiaging gene, the expression of which declines with age. Klotho may protect against neuronal oxidative damage that is induced by glutamate. The present study investigated the effects of Klotho overexpression and knockdown by an intracerebroventricular injection of a lentiviral vector that encoded murine Klotho (LV-KL) or rat Klotho short-hairpin RNA (LV-KL shRNA) on cerebral ischemia injury and the underlying anti-neuroinflammatory mechanism. The overexpression of Klotho induced by LV-KL significantly improved neurobehavioral deficits and increased the number of live neurons in the hippocampal CA1 and caudate putamen subregions 72 h after cerebral hypoperfusion that was induced by transient bilateral common carotid artery occlusion (2VO) in mice. The overexpression of Klotho significantly decreased the immunoreactivity of glial fibrillary acidic protein and ionized calcium binding adaptor molecule-1, the expression of retinoic-acid-inducible gene-I, the nuclear translocation of nuclear factor-κB, and the production of proinflammatory cytokines (tumor necrosis factor α and interleukin-6) in 2VO mice. The knockdown of Klotho mediated by LV-KL shRNA in the brain exacerbated neurological dysfunction and cerebral infarct after 22 h of reperfusion following 2 h middle cerebral artery occlusion in rats. These findings suggest that Klotho itself or enhancers of Klotho may compensate for its aging-related decline, thus providing a promising therapeutic approach for acute ischemic stroke during advanced age.
Collapse
Affiliation(s)
- Hong-Jing Zhou
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hui Li
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Meng-Qi Shi
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xiao-Na Mao
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Dong-Ling Liu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yi-Ran Chang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yu-Miao Gan
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xi Kuang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Li J, Chen S, Zhao Z, Luo Y, Hou Y, Li H, He L, Zhou L, Wu W. Effect of VEGF on Inflammatory Regulation, Neural Survival, and Functional Improvement in Rats following a Complete Spinal Cord Transection. Front Cell Neurosci 2017; 11:381. [PMID: 29238292 PMCID: PMC5712574 DOI: 10.3389/fncel.2017.00381] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/14/2017] [Indexed: 11/13/2022] Open
Abstract
After complete transection of the thoracic spinal segment, neonatal rats exhibit spontaneous locomotor recovery of hindlimbs, but this recovery is not found in adult rats after similar injury. The potential mechanism related to the difference in recovery of neonatal and adult rats remains unknown. In this study, 342 animals were analyzed. The vascular endothelial growth factor (VEGF) level in spinal segments below injury sites was significantly higher in postnatal day 1 rats (P1) compared with 28-day-old adult rats (P28) following a complete T9 transection. VEGF administration in P28 rats with T9 transection significantly improved the functional recovery; by contrast, treatment with VEGF receptor inhibitors in P1 rats with T9 transection slowed down the spontaneous functional recovery. Results showed more neurons reduced in the lumbar spinal cord and worse local neural network reorganization below injury sites in P28 rats than those in P1 rats. Transynaptic tracing with pseudorabies virus and double immunofluorescence analysis indicated that VEGF treatment in P28 rats alleviated the reduced number of neurons and improved their network reorganization. VEGF inhibition in neonates resulted in high neuronal death rate and deteriorated network reorganization. In in vivo studies, T9 transection induced less increase in the number of microglia in the spinal cord in P1 animals than P28 animals. VEGF treatment reduced the increase in microglial cells in P28 animals. VEGF administration in cultured spinal motoneurons prevented lipopolysaccharide (LPS)-induced neuronal death and facilitated neurite growth. Western blots of the samples of lumbar spinal cord after spinal transection and cultured spinal motoneurons showed a lower level of Erk1/2 phosphorylation after the injury or LPS induction compared with that in the control. The phosphorylation level increased after VEGF treatment. In conclusion, VEGF is a critical mediator involved in functional recovery after spinal transection and can be considered a potential target for clinical therapy.
Collapse
Affiliation(s)
- Jing Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
- Department of Anatomy, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Shuangxi Chen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Zhikai Zhao
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Yunhao Luo
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Yuhui Hou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Heng Li
- Department of Anatomy, University of Hong Kong, Hong Kong, Hong Kong
| | - Liumin He
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Wutian Wu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
- Department of Anatomy, University of Hong Kong, Hong Kong, Hong Kong
- Re-Stem Biotechnology Co., Ltd., Suzhou, China
| |
Collapse
|
19
|
Bhuiyan MIH, Song S, Yuan H, Begum G, Kofler J, Kahle KT, Yang SS, Lin SH, Alper SL, Subramanya AR, Sun D. WNK-Cab39-NKCC1 signaling increases the susceptibility to ischemic brain damage in hypertensive rats. J Cereb Blood Flow Metab 2017; 37:2780-2794. [PMID: 27798271 PMCID: PMC5536788 DOI: 10.1177/0271678x16675368] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
With-no-lysine kinase (WNK) and Na+-K+-2Cl- cotransporter 1 (NKCC1) are involved in the pathogenesis of hypertension. In this study, we investigated the WNK-NKCC1 signaling pathway in spontaneously hypertensive rats (SHR) and their associated susceptibility to stroke injury. Basal NKCC1 protein levels were higher in SHR than in normotensive Wistar Kyoto (WKY) rat brains. After inducing ischemic stroke, adult male WKY and SHR received either saline or NKCC1 inhibitor bumetanide (10 mg/kg/day, i.p.) starting at 3-h post-reperfusion. NKCC1 inhibition blunted the extent of ischemic infarction in SHR and improved their neurobehavioral functions. Interestingly, ischemia led to increased NKCC1 phosphorylation in SHR but not in WKY rats. Pronounced elevation of WNK1, WNK2 and WNK4 protein and downregulation of WNK3 were detected in ischemic SHR, but not in ischemic WKY rats. Upregulation of WNK-NKCC1 complex in ischemic SHR brain was associated with increased Ca2+-binding protein 39 (Cab39), without increases in Ste20-related proline alanine-rich kinase or oxidative stress-responsive kinase-1. Moreover, subacute middle cerebral artery stroke human brain autopsy exhibited increased expression of NKCC1 protein. We conclude that augmented WNK-Cab39-NKCC1 signaling in SHR is associated with an increased susceptibility to ischemic brain damage and may serve as a novel target for anti-hypertensive and anti-ischemic stroke therapy.
Collapse
Affiliation(s)
| | - Shanshan Song
- 1 Department of Neurology, University of Pittsburgh, Pittsburgh, USA
| | - Hui Yuan
- 1 Department of Neurology, University of Pittsburgh, Pittsburgh, USA
| | - Gulnaz Begum
- 1 Department of Neurology, University of Pittsburgh, Pittsburgh, USA
| | - Julia Kofler
- 2 Department of Pathology, University of Pittsburgh, Pittsburgh, USA
| | - Kristopher T Kahle
- 3 Department of Neurosurgery, Yale University School of Medicine, New Haven, USA.,4 Department of Pediatrics, Yale University School of Medicine, New Haven, USA
| | - Sung-Sen Yang
- 5 Division of Nephrology, Department of Medicine, Tri-Service General Hospital, Taipei, Taiwan.,6 Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hua Lin
- 5 Division of Nephrology, Department of Medicine, Tri-Service General Hospital, Taipei, Taiwan.,6 Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Seth L Alper
- 7 Division of Nephrology and Vascular Biology Center, Beth Israel Deaconess Medical Center, Boston, USA.,8 Department of Medicine, Harvard Medical School, Boston, USA
| | - Arohan R Subramanya
- 9 Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Dandan Sun
- 1 Department of Neurology, University of Pittsburgh, Pittsburgh, USA.,10 Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, USA
| |
Collapse
|
20
|
An in vivo study of hypoxia-inducible factor-1α signaling in ginsenoside Rg1-mediated brain repair after hypoxia/ischemia brain injury. Pediatr Res 2017; 81:120-126. [PMID: 27632778 DOI: 10.1038/pr.2016.178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/13/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hypoxia/ischemia (HI) brain injury is a common central nervous system insult in newborns. Studies have demonstrated bioactivity of ginsenoside Rg1 in increasing neural viability and promoting angiogenesis. However, there are few reports on roles of Rg1 in brain repair of neonatal HI, and the mechanisms involved are unclear. METHODS a neonatal HI model was established by a modified Rice-Vannucci model (RVM) and pups received ginsenoside Rg1 or monosialotetrahexosyl ganglioside (GM1) treatment. Neurological function and pathologic damage of rats were evaluated. Cellular apoptosis was detected with Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Immunohistochemistry for von willebrand factor (vwf) was used to label micro vessels. Expression levels of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), and cleaved caspase 3 (CC3) were detected by western blot. RESULTS Both Rg1 and GM1 reduced neurological impairment and pathologic damage after HI by enhancing neural survival. Rg1, but not GM1, could also facilitate angiogenesis after HI. These pharmacological effects of Rg1 may be attributed to regulation of expression level of VEGF and CC3 and HIF-1α signaling pathway was involved. CONCLUSION Rg1 plays a neuroprotective role in brain repair following neonatal HI, and HIF-1α is a potential target for therapeutic intervention in neonates with HI brain injury.
Collapse
|
21
|
Swaminathan A, Delage H, Chatterjee S, Belgarbi-Dutron L, Cassel R, Martinez N, Cosquer B, Kumari S, Mongelard F, Lannes B, Cassel JC, Boutillier AL, Bouvet P, Kundu TK. Transcriptional Coactivator and Chromatin Protein PC4 Is Involved in Hippocampal Neurogenesis and Spatial Memory Extinction. J Biol Chem 2016; 291:20303-14. [PMID: 27471272 DOI: 10.1074/jbc.m116.744169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Indexed: 01/12/2023] Open
Abstract
Although the elaborate combination of histone and non-histone protein complexes defines chromatin organization and hence regulates numerous nuclear processes, the role of chromatin organizing proteins remains unexplored at the organismal level. The highly abundant, multifunctional, chromatin-associated protein and transcriptional coactivator positive coactivator 4 (PC4/Sub1) is absolutely critical for life, because its absence leads to embryonic lethality. Here, we report results obtained with conditional PC4 knock-out (PC4(f/f) Nestin-Cre) mice where PC4 is knocked out specifically in the brain. Compared with the control (PC4(+/+) Nestin-Cre) mice, PC4(f/f) Nestin-Cre mice are smaller with decreased nocturnal activity but are fertile and show no motor dysfunction. Neurons in different areas of the brains of these mice show sensitivity to hypoxia/anoxia, and decreased adult neurogenesis was observed in the dentate gyrus. Interestingly, PC4(f/f) Nestin-Cre mice exhibit a severe deficit in spatial memory extinction, whereas acquisition and long term retention were unaffected. Gene expression analysis of the dorsal hippocampus of PC4(f/f) Nestin-Cre mice revealed dysregulated expression of several neural function-associated genes, and PC4 was consistently found to localize on the promoters of these genes, indicating that PC4 regulates their expression. These observations indicate that non-histone chromatin-associated proteins like PC4 play a significant role in neuronal plasticity.
Collapse
Affiliation(s)
- Amrutha Swaminathan
- From the Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore-560064, India
| | - Hélène Delage
- the Université de Lyon, Ecole Normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Snehajyoti Chatterjee
- the Laboratoire de Neurosciences Cognitives et Adaptatives, UMR7364, Université de Strasbourg, F-67000, Strasbourg, France, the UMR 7364, Laboratoire de Neurosciences Cognitives et Adaptatives, CNRS, F-67000, Strasbourg, France, and
| | | | - Raphaelle Cassel
- the Laboratoire de Neurosciences Cognitives et Adaptatives, UMR7364, Université de Strasbourg, F-67000, Strasbourg, France, the UMR 7364, Laboratoire de Neurosciences Cognitives et Adaptatives, CNRS, F-67000, Strasbourg, France, and
| | - Nicole Martinez
- the Université de Lyon, Ecole Normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Brigitte Cosquer
- the Laboratoire de Neurosciences Cognitives et Adaptatives, UMR7364, Université de Strasbourg, F-67000, Strasbourg, France
| | - Sujata Kumari
- From the Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore-560064, India
| | - Fabien Mongelard
- the Université de Lyon, Ecole Normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Béatrice Lannes
- the Département de Pathologie, Hôpital de Hautepierre, Université de Strasbourg, 67081 Strasbourg, France
| | - Jean-Christophe Cassel
- the Laboratoire de Neurosciences Cognitives et Adaptatives, UMR7364, Université de Strasbourg, F-67000, Strasbourg, France, the UMR 7364, Laboratoire de Neurosciences Cognitives et Adaptatives, CNRS, F-67000, Strasbourg, France, and
| | - Anne-Laurence Boutillier
- the Laboratoire de Neurosciences Cognitives et Adaptatives, UMR7364, Université de Strasbourg, F-67000, Strasbourg, France, the UMR 7364, Laboratoire de Neurosciences Cognitives et Adaptatives, CNRS, F-67000, Strasbourg, France, and
| | - Philippe Bouvet
- the Université de Lyon, Ecole Normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France,
| | - Tapas K Kundu
- From the Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore-560064, India,
| |
Collapse
|