1
|
Peng L, Li K, Li D, Zuo X, Zhan L, Chen M, Gong M, Sun W, Xu E. The p75 neurotrophin receptor attenuates secondary thalamic damage after cortical infarction by promoting angiogenesis. CNS Neurosci Ther 2024; 30:e14875. [PMID: 39072998 DOI: 10.1111/cns.14875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/23/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Angiogenesis is crucial in neuroprotection of secondary thalamic injury after cortical infarction. The p75 neurotrophin receptor (p75NTR) plays a key role in activating angiogenesis. However, the effects of p75NTR on angiogenesis in the thalamus after cortical infarction are largely unknown. Herein we investigate whether p75NTR facilitates angiogenesis to attenuate secondary thalamic damage via activating hypoxia-inducible factor 1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway mediated by Von Hippel-Lindau (VHL) after distal middle cerebral artery occlusion (dMCAO). METHODS The male rat model of dMCAO was established. The effects of p75NTR on the angiogenesis was evaluated using RNA-sequencing, immunohistochemistry, western blot, quantitative real-time polymerase chain reaction, magnetic resonance imaging, behavior tests, viral and pharmacological interventions. RESULTS We found that the p75NTR and vessel density were decreased in ipsilateral thalamus after dMCAO. The p75NTR-VHL interaction was reduced, which promoted the ubiquitination degradation of HIF-1α and reduced VEGF expression after dMCAO. Notably, p75NTR overexpression restrained the ubiquitination degradation of HIF-1α by inhibiting VHL-HIF-1α interaction, further promoted angiogenesis, increased cerebral blood flow of ipsilateral thalamus and improved neurological function after dMCAO. CONCLUSION For the first time, we highlighted that the enhancement of p75NTR-VHL interaction promoted angiogenesis in attenuating secondary thalamic damage after dMCAO.
Collapse
Affiliation(s)
- Linhui Peng
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kongping Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Neurology, The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dan Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xialin Zuo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixuan Zhan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Meiyan Chen
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ming Gong
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weiwen Sun
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - En Xu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Mirzahosseini G, Adam JM, Nasoohi S, El-Remessy AB, Ishrat T. Lost in Translation: Neurotrophins Biology and Function in the Neurovascular Unit. Neuroscientist 2023; 29:694-714. [PMID: 35769016 DOI: 10.1177/10738584221104982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The neurovascular unit (NVU) refers to the functional building unit of the brain and the retina, where neurons, glia, and microvasculature orchestrate to meet the demand of the retina's and brain's function. Neurotrophins (NTs) are structural families of secreted proteins and are known for exerting neurotrophic effects on neuronal differentiation, survival, neurite outgrowth, synaptic formation, and plasticity. NTs include several molecules, such as nerve growth factor, brain-derived neurotrophic factor, NT-3, NT-4, and their precursors. Furthermore, NTs are involved in signaling pathways such as inflammation, apoptosis, and angiogenesis in a nonneuronal cell type. Interestingly, NTs and the precursors can bind and activate the p75 neurotrophin receptor (p75NTR) at low and high affinity. Mature NTs bind their cognate tropomyosin/tyrosine-regulated kinase receptors, crucial for maintenance and neuronal development in the brain and retina axis. Activation of p75NTR results in neuronal apoptosis and cell death, while tropomysin receptor kinase upregulation contributes to differentiation and cell growth. Recent findings indicate that modulation of NTs and their receptors contribute to neurovascular dysfunction in the NVU. Several chronic metabolic and acute ischemic diseases affect the NVU, including diabetic and ischemic retinopathy for the retina, as well as stroke, acute encephalitis, and traumatic brain injury for the brain. This work aims to review the current evidence through published literature studying the impact of NTs and their receptors, including the p75NTR receptor, on the injured and healthy brain-retina axis.
Collapse
Affiliation(s)
- Golnoush Mirzahosseini
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Justin Mark Adam
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
3
|
Goten C, Usui S, Takashima SI, Inoue O, Yamaguchi K, Hashimuko D, Takeda Y, Nomura A, Sakata K, Kaneko S, Takamura M. Important Role of Endogenous Nerve Growth Factor Receptor in the Pathogenesis of Hypoxia-Induced Pulmonary Hypertension in Mice. Int J Mol Sci 2023; 24:1868. [PMID: 36768190 PMCID: PMC9916204 DOI: 10.3390/ijms24031868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/30/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) remains a disease with poor prognosis; thus, a new mechanism for PAH treatment is necessary. Circulating nerve growth factor receptor (Ngfr)-positive cells in peripheral blood mononuclear cells are associated with disease severity and the prognosis of PAH patients; however, the role of Ngfr in PAH is unknown. In this study, we evaluated the function of Ngfr using Ngfr gene-deletion (Ngfr-/-) mice. To elucidate the role of Ngfr in pulmonary hypertension (PH), we used Ngfr-/- mice that were exposed to chronic hypoxic conditions (10% O2) for 3 weeks. The development of hypoxia-induced PH was accelerated in Ngfr-/- mice compared to littermate controls. In contrast, the reconstitution of bone marrow (BM) in Ngfr-/- mice transplanted with wild-type BM cells improved PH. Notably, the exacerbation of PH in Ngfr-/- mice was accompanied by the upregulation of pulmonary vascular remodeling-related genes in lung tissue. In a hypoxia-induced PH model, Ngfr gene deletion resulted in PH exacerbation. This suggests that Ngfr may be a key molecule involved in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Chiaki Goten
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Soichiro Usui
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Shin-ichiro Takashima
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Oto Inoue
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Kosei Yamaguchi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Daiki Hashimuko
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Yusuke Takeda
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Ayano Nomura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Kenji Sakata
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Shuichi Kaneko
- Department of Information-Based Medicine Development, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Ishikawa, Japan
| |
Collapse
|
4
|
Morano NC, Smith RS, Danelon V, Schreiner R, Patel U, Herrera NG, Smith C, Olson SM, Laerke MK, Celikgil A, Garforth SJ, Garrett-Thomson SC, Lee FS, Hempstead BL, Almo SC. Human immunomodulatory ligand B7-1 mediates synaptic remodeling via the p75 neurotrophin receptor. J Clin Invest 2022; 132:e157002. [PMID: 36107635 PMCID: PMC9663165 DOI: 10.1172/jci157002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 09/13/2022] [Indexed: 12/30/2023] Open
Abstract
Cell surface receptors, ligands, and adhesion molecules underlie development, circuit formation, and synaptic function of the central nervous system and represent important therapeutic targets for many neuropathologies. The functional contributions of interactions between cell surface proteins of neurons and nonneuronal cells have not been fully addressed. Using an unbiased protein-protein interaction screen, we showed that the human immunomodulatory ligand B7-1 (hB7-1) interacts with the p75 neurotrophin receptor (p75NTR) and that the B7-1:p75NTR interaction is a recent evolutionary adaptation present in humans and other primates, but absent in mice, rats, and other lower mammals. The surface of hB7-1 that engages p75NTR overlaps with the hB7-1 surface involved in CTLA-4/CD28 recognition, and these molecules directly compete for binding to p75NTR. Soluble or membrane-bound hB7-1 altered dendritic morphology of cultured hippocampal neurons, with loss of the postsynaptic protein PSD95 in a p75NTR-dependent manner. Abatacept, an FDA-approved therapeutic (CTLA-4-hFc fusion) inhibited these processes. In vivo injection of hB7-1 into the murine subiculum, a hippocampal region affected in Alzheimer's disease, resulted in p75NTR-dependent pruning of dendritic spines. Here, we report the biochemical interaction between B7-1 and p75NTR, describe biological effects on neuronal morphology, and identify a therapeutic opportunity for treatment of neuroinflammatory diseases.
Collapse
Affiliation(s)
- Nicholas C. Morano
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, USA
| | - Roshelle S. Smith
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Victor Danelon
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Ryan Schreiner
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Uttsav Patel
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Natalia G. Herrera
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Carla Smith
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Steven M. Olson
- Department of Computer Science, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Michelle K. Laerke
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Alev Celikgil
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Scott J. Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Francis S. Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, New York, USA
| | - Barbara L. Hempstead
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
5
|
Ren J, Zhang S, Pan Y, Jin M, Li J, Luo Y, Sun X, Li G. Diabetic retinopathy: Involved cells, biomarkers, and treatments. Front Pharmacol 2022; 13:953691. [PMID: 36016568 PMCID: PMC9396039 DOI: 10.3389/fphar.2022.953691] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic retinopathy (DR), a leading cause of vision loss and blindness worldwide, is caused by retinal neurovascular unit dysfunction, and its cellular pathology involves at least nine kinds of retinal cells, including photoreceptors, horizontal and bipolar cells, amacrine cells, retinal ganglion cells, glial cells (Müller cells, astrocytes, and microglia), endothelial cells, pericytes, and retinal pigment epithelial cells. Its mechanism is complicated and involves loss of cells, inflammatory factor production, neovascularization, and BRB impairment. However, the mechanism has not been completely elucidated. Drug treatment for DR has been gradually advancing recently. Research on potential drug targets relies upon clear information on pathogenesis and effective biomarkers. Therefore, we reviewed the recent literature on the cellular pathology and the diagnostic and prognostic biomarkers of DR in terms of blood, protein, and clinical and preclinical drug therapy (including synthesized molecules and natural molecules). This review may provide a theoretical basis for further DR research.
Collapse
Affiliation(s)
- Jiahui Ren
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Kunming, China
| | - Shuxia Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Yunfeng Pan
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Jiaxin Li
- Yunnan Key Laboratory of Southern Medicine Utilization, Kunming, China
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- *Correspondence: Yun Luo, ; Xiaobo Sun , ; Guang Li,
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- *Correspondence: Yun Luo, ; Xiaobo Sun , ; Guang Li,
| | - Guang Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Kunming, China
- *Correspondence: Yun Luo, ; Xiaobo Sun , ; Guang Li,
| |
Collapse
|
6
|
Huang J, Tan YX, Xue LL, Du RL, Chen JJ, Chen L, Li TT, Bai X, Yang SJ, Xiong LL, Wang TH. Panax notoginseng saponin attenuates the hypoxic-ischaemic injury in neonatal rats by regulating the expression of neurotrophin factors. Eur J Neurosci 2021; 54:6304-6321. [PMID: 34405468 DOI: 10.1111/ejn.15428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/06/2021] [Accepted: 08/04/2021] [Indexed: 02/05/2023]
Abstract
Neonatal hypoxic-ischaemic (HI) injury is a serious complication of neonatal asphyxia and the leading cause of neonatal acute death and chronic neurological injury, and the effective therapeutic method is lacking to improve patients' outcomes. We reported in this study that panax notoginseng saponin (PNS) may provide a treatment option for HI. HI model was established using neonatal Sprague-Dawley rats and then intraperitoneally injected with different dosage of PNS, once a day for 7 days. Histological staining and behavioural evaluations were performed to elucidate the pathological changes and neurobehavioural variation after PNS treatment. We found PNS administration significantly reduced the infarct volume of brain tissues and improved the autonomous activities of neonatal rats, especially with higher dosage. PNS treatment at 40 mg/kg reduced neuronal damage, suppressed neuronal apoptosis and depressed astroglial reactive response. Moreover, the long-term cognitive and motor functions were also improved after PNS treatment at 40 mg/kg. Importantly, PNS treatment elevated the levels of BDNF and TrkB but decreased the expression of p75NTR both in the cortex and hippocampus of HI rats. The therapeutic efficacy of PNS might be correlated with PNS-activated BDNF/TrkB signalling and inactivation of p75NTR expression, providing a novel potential therapy for alleviating HI injury.
Collapse
Affiliation(s)
- Jin Huang
- Institute of Neuroscience, School of Preclinical Medicine, Kunming Medical University, Kunming, China
| | - Ya-Xin Tan
- Institute of Neuroscience, School of Preclinical Medicine, Kunming Medical University, Kunming, China.,Department of Pediatrics, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Lu-Lu Xue
- Animal Zoology Department, Kunming Medical University, Kunming, China
| | - Ruo-Lan Du
- Animal Zoology Department, Kunming Medical University, Kunming, China
| | - Jun-Jie Chen
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Li Chen
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting-Ting Li
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Bai
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Si-Jin Yang
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Liu-Lin Xiong
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ting-Hua Wang
- Animal Zoology Department, Kunming Medical University, Kunming, China.,Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
In vivo functions of p75 NTR: challenges and opportunities for an emerging therapeutic target. Trends Pharmacol Sci 2021; 42:772-788. [PMID: 34334250 DOI: 10.1016/j.tips.2021.06.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/31/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022]
Abstract
The p75 neurotrophin receptor (p75NTR) functions at the molecular nexus of cell death, survival, and differentiation. In addition to its contribution to neurodegenerative diseases and nervous system injuries, recent studies have revealed unanticipated roles of p75NTR in liver repair, fibrinolysis, lung fibrosis, muscle regeneration, and metabolism. Linking these various p75NTR functions more precisely to specific mechanisms marks p75NTR as an emerging candidate for therapeutic intervention in a wide range of disorders. Indeed, small molecule inhibitors of p75NTR binding to neurotrophins have shown efficacy in models of Alzheimer's disease (AD) and neurodegeneration. Here, we outline recent advances in understanding p75NTR pleiotropic functions in vivo, and propose an integrated view of p75NTR and its challenges and opportunities as a pharmacological target.
Collapse
|
8
|
Elshaer SL, Park HS, Pearson L, Hill WD, Longo FM, El-Remessy AB. Modulation of p75 NTR on Mesenchymal Stem Cells Increases Their Vascular Protection in Retinal Ischemia-Reperfusion Mouse Model. Int J Mol Sci 2021; 22:E829. [PMID: 33467640 PMCID: PMC7830385 DOI: 10.3390/ijms22020829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising therapy to improve vascular repair, yet their role in ischemic retinopathy is not fully understood. The aim of this study is to investigate the impact of modulating the neurotrophin receptor; p75NTR on the vascular protection of MSCs in an acute model of retinal ischemia/reperfusion (I/R). Wild type (WT) and p75NTR-/- mice were subjected to I/R injury by increasing intra-ocular pressure to 120 mmHg for 45 min, followed by perfusion. Murine GFP-labeled MSCs (100,000 cells/eye) were injected intravitreally 2 days post-I/R and vascular homing was assessed 1 week later. Acellular capillaries were counted using trypsin digest 10-days post-I/R. In vitro, MSC-p75NTR was modulated either genetically using siRNA or pharmacologically using the p75NTR modulator; LM11A-31, and conditioned media were co-cultured with human retinal endothelial cells (HREs) to examine the angiogenic response. Finally, visual function in mice undergoing retinal I/R and receiving LM11A-31 was assessed by visual-clue water-maze test. I/R significantly increased the number of acellular capillaries (3.2-Fold) in WT retinas, which was partially ameliorated in p75NTR-/- retinas. GFP-MSCs were successfully incorporated and engrafted into retinal vasculature 1 week post injection and normalized the number of acellular capillaries in p75NTR-/- retinas, yet ischemic WT retinas maintained a 2-Fold increase. Silencing p75NTR on GFP-MSCs coincided with a higher number of cells homing to the ischemic WT retinal vasculature and normalized the number of acellular capillaries when compared to ischemic WT retinas receiving scrambled-GFP-MSCs. In vitro, silencing p75NTR-MSCs enhanced their secretome, as evidenced by significant increases in SDF-1, VEGF and NGF release in MSCs conditioned medium; improved paracrine angiogenic response in HREs, where HREs showed enhanced migration (1.4-Fold) and tube formation (2-Fold) compared to controls. In parallel, modulating MSCs-p75NTR using LM11A-31 resulted in a similar improvement in MSCs secretome and the enhanced paracrine angiogenic potential of HREs. Further, intervention with LM11A-31 significantly mitigated the decline in visual acuity post retinal I/R injury. In conclusion, p75NTR modulation can potentiate the therapeutic potential of MSCs to harness vascular repair in ischemic retinopathy diseases.
Collapse
Affiliation(s)
- Sally L. Elshaer
- Augusta Biomedical Research Corporation, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA; (S.L.E.); (L.P.); (W.D.H.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Hang-soo Park
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA;
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| | - Laura Pearson
- Augusta Biomedical Research Corporation, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA; (S.L.E.); (L.P.); (W.D.H.)
| | - William D. Hill
- Augusta Biomedical Research Corporation, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA; (S.L.E.); (L.P.); (W.D.H.)
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA
| | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA 94304, USA;
| | - Azza B. El-Remessy
- Augusta Biomedical Research Corporation, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA; (S.L.E.); (L.P.); (W.D.H.)
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612, USA;
- Department of the Pharmacy, Doctors Hospital of Augusta, Augusta, GA 30909, USA
| |
Collapse
|
9
|
Pereira LMS, da Silva Graça Amoras E, da Silva Conde SRS, Demachki S, dos Santos EJM, Lima SS, Ishak R, Rosário Vallinoto AC. NGF (-198C > T, Ala35Val) and p75 NTR (Ser205Leu) gene mutations are associated with liver function in different histopathological profiles of the patients with chronic viral hepatitis in the Brazilian Amazon. Mol Med 2020; 26:12. [PMID: 31996124 PMCID: PMC6990582 DOI: 10.1186/s10020-019-0134-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDS Neural growth factor (NGF) is a neurotrophin that can interact with the p75NTR receptor and initiate a cascade of reactions that determines cell survival or death, and both are associated with the physiology of liver tissue. Single nucleotide polymorphisms (SNPs) in the NGF and p75NTR genes have been investigated in different pathologies; however, there are no studies that have analyzed their biological roles in the hepatic microenvironment. In the present study, we evaluated the impact of SNPs in these genes on the maintenance of liver function at different stages of inflammation and fibrosis in patients with chronic viral liver disease in the Brazilian Amazon. METHODS The SNPs -198C > T, Arg80Gln, Val72Met, Ala35Val, Ala18Ala and Ser205Leu were genotyped by real-time PCR in samples from patients with chronic viral hepatitis stratified by stage of inflammation and liver fibrosis. Histopathological, viral load (VL), liver enzyme and comorbidities data were obtained from updated medical records. Other aspects were highlighted by applied epidemiological questionnaires. RESULTS The -198C/T and Ala35Val polymorphisms in NGF were associated with changes in histopathological profiles, VL and liver enzymes. Ser205Leu polymorphism in p75NTR was associated only with changes in VL and liver enzymes. Polymorphic frequencies were variable among different ethnic populations, mainly for biologically relevant polymorphisms. A multifactorial network of interactions has been established based on genetic, virological, behavioral and biochemical aspects. CONCLUSION Mutations in the NGF (-198C > T, Ala35Val) and p75NTR (Ser205Leu) genes, within the list of multifactorial aspects, are associated with liver function in different histopathological profiles of patients with chronic viral liver disease in the Brazilian Amazon.
Collapse
Affiliation(s)
- Leonn Mendes Soares Pereira
- Virology Laboratory, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
- Postgraduate Program in Biology of Infectious and Parasitic Agents, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
| | | | | | - Sâmia Demachki
- School of Medicine, Health Science Institute, Federal University of Pará, Belém, Pará Brazil
| | - Eduardo José Melo dos Santos
- Postgraduate Program in Biology of Infectious and Parasitic Agents, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
- Laboartory of Human and Medical Genetics, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
| | - Sandra Souza Lima
- Virology Laboratory, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
| | - Ricardo Ishak
- Virology Laboratory, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
- Postgraduate Program in Biology of Infectious and Parasitic Agents, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
| | - Antonio Carlos Rosário Vallinoto
- Virology Laboratory, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
- Postgraduate Program in Biology of Infectious and Parasitic Agents, Biological Science Institute, Federal University of Pará, Belém, Pará Brazil
| |
Collapse
|
10
|
Elshaer SL, Alwhaibi A, Mohamed R, Lemtalsi T, Coucha M, Longo FM, El-Remessy AB. Modulation of the p75 neurotrophin receptor using LM11A-31 prevents diabetes-induced retinal vascular permeability in mice via inhibition of inflammation and the RhoA kinase pathway. Diabetologia 2019; 62:1488-1500. [PMID: 31073629 PMCID: PMC8808141 DOI: 10.1007/s00125-019-4885-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/28/2019] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Breakdown of the inner blood-retinal barrier (BRB) is an early event in the pathogenesis of diabetic macular oedema, that eventually leads to vision loss. We have previously shown that diabetes causes an imbalance of nerve growth factor (NGF) isoforms resulting in accumulation of its precursor proNGF and upregulation of the p75 neurotrophin receptor (p75NTR), with consequent increases in the activation of Ras homologue gene family, member A (RhoA). We also showed that genetic deletion of p75NTR in diabetes preserved the BRB and prevented inflammatory mediators in retinas. This study aims to examine the therapeutic potential of LM11A-31, a small-molecule p75NTR modulator and proNGF antagonist, in preventing diabetes-induced BRB breakdown. The study also examined the role of p75NTR/RhoA downstream signalling in mediating cell permeability. METHODS Male C57BL/6 J mice were rendered diabetic using streptozotocin injection. After 2 weeks of diabetes, mice received oral gavage of LM11A-31 (50 mg kg-1 day-1) or saline (NaCl 154 mmol/l) for an additional 4 weeks. BRB breakdown was assessed by extravasation of BSA-AlexaFluor-488. Direct effects of proNGF were examined in human retinal endothelial (HRE) cells in the presence or absence of LM11A-31 or the Rho kinase inhibitor Y-27632. RESULTS Diabetes triggered BRB breakdown and caused significant increases in circulatory and retinal TNF-α and IL-1β levels. These effects coincided with significant decreases in retinal NGF and increases in vascular endothelial growth factor and proNGF expression, as well as activation of RhoA. Interventional modulation of p75NTR activity through treatment of mouse models of diabetes with LM11A-31 significantly mitigated proNGF accumulation and preserved BRB integrity. In HRE cells, treatment with mutant proNGF (10 ng/ml) triggered increased cell permeability with marked reduction of expression of tight junction proteins, zona occludens-1 (ZO-1) and claudin-5, compared with control, independent of inflammatory mediators or cell death. Modulating p75NTR significantly inhibited proNGF-mediated RhoA activation, occludin phosphorylation (at serine 490) and cell permeability. ProNGF induced redistribution of ZO-1 in the cell wall and formation of F-actin stress fibres; these effects were mitigated by LM11A-31. CONCLUSIONS/INTERPRETATION Targeting p75NTR signalling using LM11A-31, an orally bioavailable receptor modulator, may offer an effective, safe and non-invasive therapeutic strategy for treating macular oedema, a major cause of blindness in diabetes.
Collapse
Affiliation(s)
- Sally L Elshaer
- Augusta Biomedical Research Corporation, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Abdulrahman Alwhaibi
- Augusta Biomedical Research Corporation, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Riyaz Mohamed
- Augusta Biomedical Research Corporation, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Tahira Lemtalsi
- Augusta Biomedical Research Corporation, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Maha Coucha
- Augusta Biomedical Research Corporation, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Azza B El-Remessy
- Augusta Biomedical Research Corporation, Augusta, GA, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
- Department of the Pharmacy, Doctors Hospital of Augusta, Augusta, GA, 30909, USA.
| |
Collapse
|
11
|
Zhu X, Stephens M. Large-scale genome-wide enrichment analyses identify new trait-associated genes and pathways across 31 human phenotypes. Nat Commun 2018; 9:4361. [PMID: 30341297 PMCID: PMC6195536 DOI: 10.1038/s41467-018-06805-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/29/2018] [Indexed: 12/27/2022] Open
Abstract
Genome-wide association studies (GWAS) aim to identify genetic factors associated with phenotypes. Standard analyses test variants for associations individually. However, variant-level associations are hard to identify and can be difficult to interpret biologically. Enrichment analyses help address both problems by targeting sets of biologically related variants. Here we introduce a new model-based enrichment method that requires only GWAS summary statistics. Applying this method to interrogate 4,026 gene sets in 31 human phenotypes identifies many previously-unreported enrichments, including enrichments of endochondral ossification pathway for height, NFAT-dependent transcription pathway for rheumatoid arthritis, brain-related genes for coronary artery disease, and liver-related genes for Alzheimer’s disease. A key feature of our method is that inferred enrichments automatically help identify new trait-associated genes. For example, accounting for enrichment in lipid transport genes highlights association between MTTP and low-density lipoprotein levels, whereas conventional analyses of the same data found no significant variants near this gene. In genome-wide association studies, variant-level associations are hard to identify and can be difficult to interpret biologically. Here, the authors develop a new model-based enrichment analysis method, and apply it to identify new associated genes, pathways and tissues across 31 human phenotypes.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Statistics, Stanford University, Stanford, 94305, CA, USA. .,Department of Statistics, The University of Chicago, Chicago, 60637, IL, USA.
| | - Matthew Stephens
- Department of Statistics, The University of Chicago, Chicago, 60637, IL, USA. .,Department of Human Genetics, The University of Chicago, Chicago, 60637, IL, USA.
| |
Collapse
|
12
|
Chen Y, Hou Y, Yang J, Du R, Chen C, Chen F, Wang H, Ge R, Chen J. P75 Involved in the Ubiquitination of α-synuclein in Rotenone-based Parkinson’s Disease Models. Neuroscience 2018; 388:367-373. [DOI: 10.1016/j.neuroscience.2018.07.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 11/29/2022]
|
13
|
Elshaer SL, El-Remessy AB. Deletion of p75 NTR prevents vaso-obliteration and retinal neovascularization via activation of Trk- A receptor in ischemic retinopathy model. Sci Rep 2018; 8:12490. [PMID: 30131506 PMCID: PMC6104090 DOI: 10.1038/s41598-018-30029-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023] Open
Abstract
Ischemic retinopathy is characterized by ischemia followed by retinal neovascularization (RNV) resulting in visual impairment. Given the role of neuron-secreted growth factors in regulating angiogenesis, we examined how genetic deletion of the neurotrophin receptor; p75NTR can overcome retinal ischemia using oxygen-induced retinopathy (OIR) mouse model. Wildtype (WT) or p75NTR-/- mice pups were subjected to hyperoxia (70% O2, p7-p12) then returned to normal air (relative hypoxia, p12-p17). Vascular alterations were assessed at p12 and p17 time-points. Deletion of p75NTR prevented hyperoxia-associated central vascular cell death (p12) and hypoxia-associated RNV and enhanced central vascular repair (p17). Decreased expression of apoptotic markers; preserved Akt survival signal decreased proNGF were also observed at p12. During hypoxia, deletion of p75NTR maintained VEGF and VEGFR2 activation and restored NGF/proNGF and BDNF/proBDNF levels. Deletion of p75NTR coincided with significant increases in expression and activation of NGF survival receptor, TrkA at basal and hyperoxic condition. Pharmacological inhibition of TrkA using compound K-252a (0.5 μg 1 μl-1/eye) resulted in 2-fold increase in pathological RNV and 1.34-fold increase in central vascular cell death in p75NTR-/- pups. In conclusion, deletion of p75NTR protected against retinal ischemia and prevented RNV, in part, through restoring neurotrophic support and activating TrkA receptor.
Collapse
Affiliation(s)
- Sally L Elshaer
- Augusta Biomedical Research Corporation, Augusta, GA, 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
- Ophthalmology Department, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38163, USA
| | - Azza B El-Remessy
- Augusta Biomedical Research Corporation, Augusta, GA, 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
| |
Collapse
|
14
|
Association assessment of Nerve growth factor gene promoter polymorphism and its expression status with susceptibility to coronary artery disease. Meta Gene 2018. [DOI: 10.1016/j.mgene.2017.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
15
|
Mohamed R, Shanab AY, El Remessy AB. Deletion of the Neurotrophin Receptor p75 NTR Prevents Diabetes-Induced Retinal Acellular Capillaries in Streptozotocin-Induced Mouse Diabetic Model. ACTA ACUST UNITED AC 2017; 4. [PMID: 29658956 DOI: 10.15406/jdmdc.2017.04.00129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Diabetic retinopathy is characterized by early stage of retinal neuro-inflammation that triggers development of acellular capillaries and a late stage of pathological neovascularization. Due to limited treatment options, there is a pressing need to develop new therapeutics. Our group discovered that diabetes-impaired processing of the nerve growth factor precursor (proNGF) resulting in its accumulation and its receptor p75NTR. Here, we examine the protective effects of modulating p75NTR in experimental model of diabetic retinopathy. Diabetes was induced using streptozotocin in both wild type (WT) and p75NTR knockout (p75KO) mice. Retinal inflammation and microvascular dysfunction were assessed. Western blot analysis was performed to assess expression of apoptotic and inflammatory markers and levels of the neurotrophin, p75NTR and ephrin-B2. Deletion of p75NTR did not alter body weight or diabetes status compared to WT mice. In WT-mice, diabetes triggered retinal inflammation, significant decrease in pericyte count and marked increase in development of occluded (acellular) capillary formation after 24-weeks. Deletion of p75NTR prevented acellular capillary, restored pericyte count, and inhibited the retinal Ephrin-B2, activation of the stress-kinase JNK and apoptotic marker cleaved caspase-3 in the diabetic retina. Deletion of p75NTR reduced retinal inflammation, and proNGF expression. These effects coincided with increased NGF level and TrkA activation in the diabetic retina. Targeting p75NTR using genetic approach protected the retina from the impact of long-term diabetes in mediating microvascular degeneration and maintains the balance of NGF/proNGF level. Together, these results provide rationale that targeting p75NTR may offer novel and effective therapeutic strategy to combat diabetic retinopathy.
Collapse
Affiliation(s)
- Riyaz Mohamed
- Charlie Norwood Veterans Affairs Medical Center, USA
| | | | - Azza B El Remessy
- Charlie Norwood Veterans Affairs Medical Center, USA.,Augusta Biomedical Research Foundation, USA
| |
Collapse
|
16
|
Inducible overexpression of endothelial proNGF as a mouse model to study microvascular dysfunction. Biochim Biophys Acta Mol Basis Dis 2017; 1864:746-757. [PMID: 29253516 DOI: 10.1016/j.bbadis.2017.12.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022]
Abstract
Impaired maturation of nerve growth factor precursor (proNGF) and its accumulation has been reported in several neurodegenerative diseases, myocardial infarction and diabetes. To elucidate the direct impact of proNGF accumulation identified the need to create a transgenic model that can express fully mutated cleavage-resistant proNGF. Using Cre-Lox technology, we developed an inducible endothelial-specific proNGF transgenic mouse (proNGFLoxp) that overexpresses GFP-conjugated cleavage-resistant proNGF123 when crossed with VE-cadherin-CreERT2 (Cre). Expression of proNGF, inflammatory mediators, NGF and VEGF was evaluated by PCR, Western blot and immunohistochemistry. EC-proNGF overexpression was confirmed using colocalization of anti-proNGF within retinal vasculature. EC-proNGF did not cause retinal neurotoxicity or marked glial activation at 4-weeks. Microvascular preparation from Cre-proNGF mice showed significant imbalance of proNGF/NGF ratio, enhanced expression of TNF-α and p75NTR, and tendency to impair TrkA phosphorylation compared to controls. EC-proNGF overexpression triggered mRNA expression of p75NTR and inflammatory mediators in both retina and renal cortex compared to controls. EC-proNGF expression induced vascular permeability including breakdown of BRB and albuminuria in the kidney without affecting VEGF level at 4-weeks. Histopathological changes were assessed after 8-weeks and the results showed that EC-proNGF triggered formation of occluded (acellular) capillaries, hall mark of retinal ischemia. EC-proNGF resulted in glomerular enlargement and kidney fibrosis, hall mark of renal dysfunction. We have successfully created an inducible mouse model that can dissect the contribution of autocrine direct action of cleavage-resistant proNGF on systemic microvascular abnormalities in both retina and kidney, major targets for microvascular complication.
Collapse
|