1
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
2
|
Chen G, Wang Y, Liu X, Liu F. Enhancing the effects of curcumin on oxidative stress injury in brain vascular endothelial cells using lactoferrin peptide nano-micelles: antioxidant activity and mechanism. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:372-381. [PMID: 39210730 DOI: 10.1002/jsfa.13836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 07/19/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Curcumin is widely known for its antioxidant and anti-inflammatory properties, but its mechanism of action in mitigating oxidative stress injury in brain vascular endothelial cells remains unclear. Due to the poor bioavailability of curcumin, it is challenging to achieve effective concentrations at the target sites. Nano-micelles are known for their ability to improve the solubility, stability, and bioavailability of hydrophobic compounds like curcumin. This study investigated the effects and mechanisms of free curcumin and curcumin embedded in nano-micelles (M(Cur)) on oxidative stress-induced injury in bEnd.3 cells. RESULTS At a protective concentration of 10 μg mL-1, micellar curcumin was better able to recover the morphology of bEnd.3 cells under oxidative stress while increasing cell viability, restoring mitochondrial membrane electrical potential, and effectively inhibiting reactive oxygen species generation with a positive cell rate of 2.21%. These results indicate that curcumin significantly improves H2O2-induced oxidative stress damage in endothelial cells by maintaining the cellular antioxidant balance. CONCLUSION This study adds to knowledge regarding the role of nano-micelles in curcumin intervention for endothelial cell oxidative damage and provides insights for the development of curcumin-based dietary supplements. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Guipan Chen
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| | - Yiyang Wang
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Xianyang, China
| |
Collapse
|
3
|
Li G, Ma Y, Zhang S, Lin W, Yao X, Zhou Y, Zhao Y, Rao Q, Qu Y, Gao Y, Chen L, Zhang Y, Han F, Sun M, Zhao C. A mechanistic systems biology model of brain microvascular endothelial cell signaling reveals dynamic pathway-based therapeutic targets for brain ischemia. Redox Biol 2024; 78:103415. [PMID: 39520909 PMCID: PMC11584692 DOI: 10.1016/j.redox.2024.103415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Ischemic stroke is a significant threat to human health. Currently, there is a lack of effective treatments for stroke, and progress in new neuron-centered drug target development is relatively slow. On the other hand, studies have demonstrated that brain microvascular endothelial cells (BMECs) are crucial components of the neurovascular unit and play pivotal roles in ischemic stroke progression. To better understand the complex multifaceted roles of BMECs in the regulation of ischemic stroke pathophysiology and facilitate BMEC-based drug target discovery, we utilized a transcriptomics-informed systems biology modeling approach and constructed a mechanism-based computational multipathway model to systematically investigate BMEC function and its modulatory potential. Extensive multilevel data regarding complex BMEC pathway signal transduction and biomarker expression under various pathophysiological conditions were used for quantitative model calibration and validation, and we generated dynamic BMEC phenotype maps in response to various stroke-related stimuli to identify potential determinants of BMEC fate under stress conditions. Through high-throughput model sensitivity analyses and virtual target perturbations in model-based single cells, our model predicted that targeting succinate could effectively reverse the detrimental cell phenotype of BMECs under oxygen and glucose deprivation/reoxygenation, a condition that mimics stroke pathogenesis, and we experimentally validated the utility of this new target in terms of regulating inflammatory factor production, free radical generation and tight junction protection in vitro and in vivo. Our work is the first that complementarily couples transcriptomic analysis with mechanistic systems-level pathway modeling in the study of BMEC function and endothelium-based therapeutic targets in ischemic stroke.
Collapse
Affiliation(s)
- Geli Li
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; Gusu School, Nanjing Medical University, 215000, Suzhou, China
| | - Yuchen Ma
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Sujie Zhang
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Wen Lin
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Xinyi Yao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yating Zhou
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yanyong Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Qi Rao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuchen Qu
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, 210000, Nanjing, China
| | - Lianmin Chen
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 21205, Baltimore, USA
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China.
| | - Meiling Sun
- School of Basic Medical Sciences, Nanjing Medical University, 210000, Nanjing, China.
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China.
| |
Collapse
|
4
|
Zhang Q, Zhang X, Zhang J, Yu N. Peripheral blood diagnostic markers of chronic cerebral hypoperfusion. Am J Transl Res 2024; 16:2973-2981. [PMID: 39114695 PMCID: PMC11301487 DOI: 10.62347/orsn1228] [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: 02/20/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE To investigate the efficacy of ischemia-modified albumin (IMA), lipoprotein-associated phospholipase A2 (Lp-PLA2), brain-derived neurotrophic factor (BDNF), and visinin-like protein-1 (VILIP-1) in diagnosing chronic cerebral hypoperfusion (CCH). METHODS This retrospective study included 84 patients with suspected chronic cerebral ischemia admitted to Sichuan Provincial People's Hospital between February 2021 and April 2022. Arterial spin labeling (ASL) imaging and biological examinations were performed. According to the ASL perfusion imaging patterns, the patients were divided into a CCH group (n = 55) and a non-CCH group (n = 29). Serum markers of the two groups were compared, and correlation analysis was conducted between ischemic marker levels and cerebral blood flow (CBF) in the ischemic region, as measured by ASL. Receiver operating characteristic (ROC) curve analysis was used to evaluate the efficacy of each marker for diagnosing chronic cerebral ischemia. The Delong test was used to compare AUC size between groups. RESULTS Compared to the non-CCH group, the CCH group exhibited higher IMA levels and lower BDNF concentrations (P < 0.05). However, VILIP-1 and Lp-PLA2 concentrations were not significantly different between the two groups (P > 0.05). Moreover, IMA and BDNF levels were not correlated with CBF in the hypoperfused area. ROC curve analysis demonstrated that the cut-off values of 24.2915 U/mL and 6.714 ng/L for IMA and BDNF achieved a sensitivity of 83.6% and 41.8% and a specificity of 62.1% and 93.1%, respectively. Lastly, the areas under the curve for IMA and BDNF were 0.738 (95% confidence interval [CI], 0.627-0.848) and 0.631 (95% CI, 0.512-0.751), respectively. CONCLUSION IMA and BDNF may have clinical value in the diagnosis of CCH.
Collapse
Affiliation(s)
- Qiong Zhang
- Clinical School of Medicine, Southwest Medical UniversityLuzhou 646000, Sichuan, China
- Jiangyou City People’s HospitalJiangyou 621700, Sichuan, China
| | - Xin Zhang
- Department of Internal Medicine-Neurology, Chengdu 363 Hospital Affiliated to Southwest Medical UniversityChengdu 610041, Sichuan, China
| | - Jing Zhang
- Chengdu Sixth People’s HospitalChengdu 610051, Sichuan, China
- Clinical Medical School, University of Electronic Science and Technology of ChinaChengdu 610054, Sichuan, China
| | - Nengwei Yu
- Clinical School of Medicine, Southwest Medical UniversityLuzhou 646000, Sichuan, China
- Clinical Medical School, University of Electronic Science and Technology of ChinaChengdu 610054, Sichuan, China
- Department of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s HospitalChengdu 610072, Sichuan, China
| |
Collapse
|
5
|
Raslan AA, Pham TX, Lee J, Kontodimas K, Tilston-Lunel A, Schmottlach J, Hong J, Dinc T, Bujor AM, Caporarello N, Thiriot A, von Andrian UH, Huang SK, Nicosia RF, Trojanowska M, Varelas X, Ligresti G. Lung injury-induced activated endothelial cell states persist in aging-associated progressive fibrosis. Nat Commun 2024; 15:5449. [PMID: 38937456 PMCID: PMC11211333 DOI: 10.1038/s41467-024-49545-x] [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: 02/08/2023] [Accepted: 06/04/2024] [Indexed: 06/29/2024] Open
Abstract
Progressive lung fibrosis is associated with poorly understood aging-related endothelial cell dysfunction. To gain insight into endothelial cell alterations in lung fibrosis we performed single cell RNA-sequencing of bleomycin-injured lungs from young and aged mice. Analysis reveals activated cell states enriched for hypoxia, glycolysis and YAP/TAZ activity in ACKR1+ venous and TrkB+ capillary endothelial cells. Endothelial cell activation is prevalent in lungs of aged mice and can also be detected in human fibrotic lungs. Longitudinal single cell RNA-sequencing combined with lineage tracing demonstrate that endothelial activation resolves in young mouse lungs but persists in aged ones, indicating a failure of the aged vasculature to return to quiescence. Genes associated with activated lung endothelial cells states in vivo can be induced in vitro by activating YAP/TAZ. YAP/TAZ also cooperate with BDNF, a TrkB ligand that is reduced in fibrotic lungs, to promote capillary morphogenesis. These findings offer insights into aging-related lung endothelial cell dysfunction that may contribute to defective lung injury repair and persistent fibrosis.
Collapse
Affiliation(s)
- Ahmed A Raslan
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Tho X Pham
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jisu Lee
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Konstantinos Kontodimas
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jillian Schmottlach
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jeongmin Hong
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Taha Dinc
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andreea M Bujor
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | | | - Aude Thiriot
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Trojanowska
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| | - Giovanni Ligresti
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
6
|
Berezin OO, Berezina TA, Hoppe UC, Lichtenauer M, Berezin AE. Diagnostic and predictive abilities of myokines in patients with heart failure. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:45-98. [PMID: 39059994 DOI: 10.1016/bs.apcsb.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Myokines are defined as a heterogenic group of numerous cytokines, peptides and metabolic derivates, which are expressed, synthesized, produced, and released by skeletal myocytes and myocardial cells and exert either auto- and paracrine, or endocrine effects. Previous studies revealed that myokines play a pivotal role in mutual communications between skeletal muscles, myocardium and remote organs, such as brain, vasculature, bone, liver, pancreas, white adipose tissue, gut, and skin. Despite several myokines exert complete divorced biological effects mainly in regulation of skeletal muscle hypertrophy, residential cells differentiation, neovascularization/angiogenesis, vascular integrity, endothelial function, inflammation and apoptosis/necrosis, attenuating ischemia/hypoxia and tissue protection, tumor growth and malignance, for other occasions, their predominant effects affect energy homeostasis, glucose and lipid metabolism, adiposity, muscle training adaptation and food behavior. Last decade had been identified 250 more myokines, which have been investigating for many years further as either biomarkers or targets for heart failure management. However, only few myokines have been allocated to a promising tool for monitoring adverse cardiac remodeling, ischemia/hypoxia-related target-organ dysfunction, microvascular inflammation, sarcopenia/myopathy and prediction for poor clinical outcomes among patients with HF. This we concentrate on some most plausible myokines, such as myostatin, myonectin, brain-derived neurotrophic factor, muslin, fibroblast growth factor 21, irisin, leukemia inhibitory factor, developmental endothelial locus-1, interleukin-6, nerve growth factor and insulin-like growth factor-1, which are suggested to be useful biomarkers for HF development and progression.
Collapse
Affiliation(s)
- Oleksandr O Berezin
- Luzerner Psychiatrie AG, Department of Senior Psychiatrie, St. Urban, Switzerland
| | - Tetiana A Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, Zaporozhye, Ukraine
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
7
|
Kim BS, Kim JU, Lee J, Ryu KM, Kim SH, Hwang NS. Decellularized brain extracellular matrix based NGF-releasing cryogel for brain tissue engineering in traumatic brain injury. J Control Release 2024; 368:140-156. [PMID: 38373473 DOI: 10.1016/j.jconrel.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Traumatic brain injuries(TBI) pose significant challenges to human health, specifically neurological disorders and related motor activities. After TBI, the injured neuronal tissue is known for hardly regenerated and recovered to their normal neuron physiology and tissue compositions. For this reason, tissue engineering strategies that promote neuronal regeneration have gained increasing attention. This study explored the development of a novel neural tissue regeneration cryogel by combining brain-derived decellularized extracellular matrix (ECM) with heparin sulfate crosslinking that can perform nerve growth factor (NGF) release ability. Morphological and mechanical characterizations of the cryogels were performed to assess their suitability as a neural regeneration platform. After that, the heparin concnentration dependent effects of varying NGF concentrations on cryogel were investigated for their controlled release and impact on neuronal cell differentiation. The results revealed a direct correlation between the concentration of released NGF and the heparin sulfate ratio in cryogel, indicating that the cryogel can be tailored to carry higher loads of NGF with heparin concentration in cryogel that induced higher neuronal cell differentiation ratio. Furthermore, the study evaluated the NGF loaded cryogels on neuronal cell proliferation and brain tissue regeneration in vivo. The in vivo results suggested that the NGF loaded brain ECM derived cryogel significantly affects the regeneration of brain tissue. Overall, this research contributes to the development of advanced neural tissue engineering strategies and provides valuable insights into the design of regenerative cryogels that can be customized for specific therapeutic applications.
Collapse
Affiliation(s)
- Beom-Seok Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Uk Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaewoo Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Min Ryu
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Nathaniel S Hwang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea; Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
8
|
Benítez-Castañeda A, Anaya-Martínez V, Espadas-Alvarez ADJ, Gutierrez-Váldez AL, Razgado-Hernández LF, Reyna-Velazquez PE, Quintero-Macias L, Martínez-Fong D, Florán-Garduño B, Aceves J. Transfection of the BDNF Gene in the Surviving Dopamine Neurons in Conjunction with Continuous Administration of Pramipexole Restores Normal Motor Behavior in a Bilateral Rat Model of Parkinson's Disease. PARKINSON'S DISEASE 2024; 2024:3885451. [PMID: 38419644 PMCID: PMC10901579 DOI: 10.1155/2024/3885451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
In Parkinson's disease (PD), progressive degeneration of nigrostriatal innervation leads to atrophy and loss of dendritic spines of striatal medium spiny neurons (MSNs). The loss disrupts corticostriatal transmission, impairs motor behavior, and produces nonmotor symptoms. Nigral neurons express brain-derived neurotropic factor (BDNF) and dopamine D3 receptors, both protecting the dopamine neurons and the spines of MSNs. To restore motor and nonmotor symptoms to normality, we assessed a combined therapy in a bilateral rat Parkinson's model, with only 30% of surviving neurons. The preferential D3 agonist pramipexole (PPX) was infused for four ½ months via mini-osmotic pumps and one month after PPX initiation; the BDNF-gene was transfected into the surviving nigral cells using the nonviral transfection NTS-polyplex vector. Overexpression of the BDNF-gene associated with continuous PPX infusion restored motor coordination, balance, normal gait, and working memory. Recovery was also related to the restoration of the average number of dendritic spines of the striatal projection neurons and the number of TH-positive neurons of the substantia nigra and ventral tegmental area. These positive results could pave the way for further clinical research into this promising therapy.
Collapse
Affiliation(s)
- Alina Benítez-Castañeda
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | | | | | | | | | | | - Liz Quintero-Macias
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Daniel Martínez-Fong
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Benjamín Florán-Garduño
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Jorge Aceves
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| |
Collapse
|
9
|
Leger C, Quirié A, Méloux A, Fontanier E, Chaney R, Basset C, Lemaire S, Garnier P, Prigent-Tessier A. Impact of Exercise Intensity on Cerebral BDNF Levels: Role of FNDC5/Irisin. Int J Mol Sci 2024; 25:1213. [PMID: 38279218 PMCID: PMC10816613 DOI: 10.3390/ijms25021213] [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: 12/22/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
The positive effects of physical exercise (EX) are well known to be mediated by cerebral BDNF (brain-derived neurotrophic factor), a neurotrophin involved in learning and memory, the expression of which could be induced by circulating irisin, a peptide derived from Fibronectin type III domain-containing protein 5 (FNDC5) produced by skeletal muscle contraction. While the influence of EX modalities on cerebral BDNF expression was characterized, their effect on muscle FNDC5/Irisin expression and circulating irisin levels remains to be explored. The present study involved Wistar rats divided into four experimental groups: sedentary (SED), low- (40% of maximal aerobic speed, MAS), intermediate- (50% of MAS) and high- (70% of MAS) intensities of treadmill EX (30 min/day, 7 days). Soleus (SOL) versus gastrocnemius (GAS) FNDC5 and hippocampal BDNF expressions were evaluated by Western blotting. Additionally, muscular FNDC5/Irisin localization and serum/hippocampal irisin levels were studied by immunofluorescence and ELISA, respectively. Our findings revealed that (1) serum irisin and hippocampal BDNF levels vary with EX intensity, showing a threshold intensity at 50% of MAS; (2) hippocampal BDNF levels positively correlate with serum irisin but not with hippocampal FNDC5/Irisin; and (3) GAS, in response to EX intensity, overexpresses FNDC5/Irisin in type II muscle fibers. Altogether, peripheral FNDC5/Irisin levels likely explain EX-dependent hippocampal BDNF expression.
Collapse
Affiliation(s)
- Clémence Leger
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
| | - Aurore Quirié
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
| | - Alexandre Méloux
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
| | - Estelle Fontanier
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
| | - Rémi Chaney
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
| | - Christelle Basset
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
| | - Stéphanie Lemaire
- Centre Hospitalier Universitaire Dijon, Service de Biochimie Spécialisée, F-21000 Dijon, France;
| | - Philippe Garnier
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
- Département Génie Biologique, Institut Universitaire et Technologique, F-21000 Dijon, France
| | - Anne Prigent-Tessier
- Inserm Unité Mixte de Recherche 1093-Cognition, Action & Plasticité Sensorimotrice, Unité de Formation et de Recherche Sciences de Santé, Université de Bourgogne, F-21000 Dijon, France; (C.L.); (A.Q.); (A.M.); (E.F.); (R.C.); (C.B.); (P.G.)
| |
Collapse
|
10
|
Cefis M, Chaney R, Wirtz J, Méloux A, Quirié A, Leger C, Prigent-Tessier A, Garnier P. Molecular mechanisms underlying physical exercise-induced brain BDNF overproduction. Front Mol Neurosci 2023; 16:1275924. [PMID: 37868812 PMCID: PMC10585026 DOI: 10.3389/fnmol.2023.1275924] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Accumulating evidence supports that physical exercise (EX) is the most effective non-pharmacological strategy to improve brain health. EX prevents cognitive decline associated with age and decreases the risk of developing neurodegenerative diseases and psychiatric disorders. These positive effects of EX can be attributed to an increase in neurogenesis and neuroplastic processes, leading to learning and memory improvement. At the molecular level, there is a solid consensus to involve the neurotrophin brain-derived neurotrophic factor (BDNF) as the crucial molecule for positive EX effects on the brain. However, even though EX incontestably leads to beneficial processes through BDNF expression, cellular sources and molecular mechanisms underlying EX-induced cerebral BDNF overproduction are still being elucidated. In this context, the present review offers a summary of the different molecular mechanisms involved in brain's response to EX, with a specific focus on BDNF. It aims to provide a cohesive overview of the three main mechanisms leading to EX-induced brain BDNF production: the neuronal-dependent overexpression, the elevation of cerebral blood flow (hemodynamic hypothesis), and the exerkine signaling emanating from peripheral tissues (humoral response). By shedding light on these intricate pathways, this review seeks to contribute to the ongoing elucidation of the relationship between EX and cerebral BDNF expression, offering valuable insights into the potential therapeutic implications for brain health enhancement.
Collapse
Affiliation(s)
- Marina Cefis
- Département des Sciences de l’Activité Physique, Faculté des Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Remi Chaney
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Julien Wirtz
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Alexandre Méloux
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Clémence Leger
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université de Bourgogne, UFR des Sciences de Santé, Dijon, France
- Département Génie Biologique, Institut Universitaire de Technologie, Dijon, France
| |
Collapse
|
11
|
Kim M, Lee J, Cai L, Choi H, Oh D, Jawad A, Hyun SH. Neurotrophin-4 promotes the specification of trophectoderm lineage after parthenogenetic activation and enhances porcine early embryonic development. Front Cell Dev Biol 2023; 11:1194596. [PMID: 37519302 PMCID: PMC10373506 DOI: 10.3389/fcell.2023.1194596] [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: 03/27/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Neurotrophin-4 (NT-4), a neurotrophic factor, appears to affect early embryonic development because it is secreted not only by neurons but also by oviductal and uterine epithelial cells. However, no studies have characterized the effects of NT-4 on early embryonic development in pigs. In this study, we applied the experimental model of parthenogenetic-activation (PA)-derived embryos. Herein, we investigated the effect of NT-4 supplementation during the in vitro culture (IVC) of embryos, analyzed the transcription levels of specific genes, and outlined the first cell lineage specification for porcine PA-derived blastocysts. We confirmed that NT-4 and its receptor proteins were localized in both the inner cell mass (ICM) and trophectoderm (TE) in porcine blastocysts. Across different concentrations (0, 1, 10, and 100 ng/mL) of NT-4 supplementation, the optimal concentration of NT-4 to improve the developmental competence of porcine parthenotes was 10 ng/mL. NT-4 supplementation during porcine IVC significantly (p < 0.05) increased the proportion of TE cells by inducing the transcription of TE lineage markers (CDX2, PPAG3, and GATA3 transcripts). NT-4 also reduced blastocyst apoptosis by regulating the transcription of apoptosis-related genes (BAX and BCL2L1 transcripts) and improved blastocyst quality via the interaction of neurotrophin-, Hippo-yes-associated protein (Hippo-YAP) and mitogen-activated protein kinase/extracellular regulated kinase (MAPK/ERK) pathway. Additionally, NT-4 supplementation during IVC significantly (p < 0.05) increased YAP1 transcript levels and significantly (p < 0.01) decreased LATS2 transcript levels, respectively, in the porcine PA-derived blastocysts. We also confirmed through fluorescence intensity that the YAP1 protein was significantly (p < 0.001) increased in the NT-4-treated blastocysts compared with that in the control. NT-4 also promoted differentiation into the TE lineage rather than into the ICM lineage during porcine early embryonic development. In conclusion, 10 ng/mL NT-4 supplementation enhanced blastocyst quality by regulating the apoptosis- and TE lineage specification-related genes and interacting with neurotrophin-, Hippo-YAP-, and MAPK/ERK signaling pathway during porcine in vitro embryo development.
Collapse
Affiliation(s)
- Mirae Kim
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Joohyeong Lee
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Lian Cai
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyerin Choi
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Dongjin Oh
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Ali Jawad
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Sang-Hwan Hyun
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
12
|
Pisani A, Paciello F, Del Vecchio V, Malesci R, De Corso E, Cantone E, Fetoni AR. The Role of BDNF as a Biomarker in Cognitive and Sensory Neurodegeneration. J Pers Med 2023; 13:jpm13040652. [PMID: 37109038 PMCID: PMC10140880 DOI: 10.3390/jpm13040652] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has a crucial function in the central nervous system and in sensory structures including olfactory and auditory systems. Many studies have highlighted the protective effects of BDNF in the brain, showing how it can promote neuronal growth and survival and modulate synaptic plasticity. On the other hand, conflicting data about BDNF expression and functions in the cochlear and in olfactory structures have been reported. Several clinical and experimental research studies showed alterations in BDNF levels in neurodegenerative diseases affecting the central and peripheral nervous system, suggesting that BDNF can be a promising biomarker in most neurodegenerative conditions, including Alzheimer's disease, shearing loss, or olfactory impairment. Here, we summarize current research concerning BDNF functions in brain and in sensory domains (olfaction and hearing), focusing on the effects of the BDNF/TrkB signalling pathway activation in both physiological and pathological conditions. Finally, we review significant studies highlighting the possibility to target BDNF as a biomarker in early diagnosis of sensory and cognitive neurodegeneration, opening new opportunities to develop effective therapeutic strategies aimed to counteract neurodegeneration.
Collapse
Affiliation(s)
- Anna Pisani
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Del Vecchio
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Malesci
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Eugenio De Corso
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Elena Cantone
- Department of Neuroscience, Reproductive Sciences and Dentistry-ENT Section, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Rita Fetoni
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
13
|
Arutjunyan AV, Kerkeshko GO, Milyutina YP, Shcherbitskaia AD, Zalozniaia IV, Mikhel AV, Inozemtseva DB, Vasilev DS, Kovalenko AA, Kogan IY. Imbalance of Angiogenic and Growth Factors in Placenta in Maternal Hyperhomocysteinemia. BIOCHEMISTRY (MOSCOW) 2023; 88:262-279. [PMID: 37072327 DOI: 10.1134/s0006297923020098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Numerous studies have shown that various adverse factors of different nature and action mechanisms have similar negative influence on placental angiogenesis, resulting in insufficiency of placental blood supply. One of the risk factors for pregnancy complications with placental etiology is an increased level of homocysteine in the blood of pregnant women. However, the effect of hyperhomocysteinemia (HHcy) on the development of the placenta and, in particular, on the formation of its vascular network is at present poorly understood. The aim of this work was to study the effect of maternal HHcy on the expression of angiogenic and growth factors (VEGF-A, MMP-2, VEGF-B, BDNF, NGF), as well as their receptors (VEGFR-2, TrkB, p75NTR), in the rat placenta. The effects of HHcy were studied in the morphologically and functionally different maternal and fetal parts of the placenta on the 14th and 20th day of pregnancy. The maternal HHcy caused increase in the levels of oxidative stress and apoptosis markers accompanied by an imbalance of the studied angiogenic and growth factors in the maternal and/or fetal part of the placenta. The influence of maternal HHcy in most cases manifested in a decrease in the protein content (VEGF-A), enzymatic activity (MMP-2), gene expression (VEGFB, NGF, TRKB), and accumulation of precursor form (proBDNF) of the investigated factors. In some cases, the effects of HHcy differed depending on the placental part and stage of development. The influence of maternal HHcy on signaling pathways and processes controlled by the studied angiogenic and growth factors could lead to incomplete development of the placental vasculature and decrease in the placental transport, resulting in fetal growth restriction and impaired fetal brain development.
Collapse
Affiliation(s)
- Alexander V Arutjunyan
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia.
| | - Gleb O Kerkeshko
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Yulia P Milyutina
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
- St. Petersburg State Pediatric Medical University, Russian Ministry of Health, St. Petersburg, 194100, Russia
| | - Anastasiia D Shcherbitskaia
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Irina V Zalozniaia
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Anastasiia V Mikhel
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Daria B Inozemtseva
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Dmitrii S Vasilev
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Anna A Kovalenko
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Igor Yu Kogan
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| |
Collapse
|
14
|
Mehta RI, Mehta RI. The Vascular-Immune Hypothesis of Alzheimer's Disease. Biomedicines 2023; 11:408. [PMID: 36830944 PMCID: PMC9953491 DOI: 10.3390/biomedicines11020408] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating and irreversible neurodegenerative disorder with unknown etiology. While its cause is unclear, a number of theories have been proposed to explain the pathogenesis of AD. In large part, these have centered around potential causes for intracerebral accumulation of beta-amyloid (βA) and tau aggregates. Yet, persons with AD dementia often exhibit autopsy evidence of mixed brain pathologies including a myriad of vascular changes, vascular brain injuries, complex brain inflammation, and mixed protein inclusions in addition to hallmark neuropathologic lesions of AD, namely insoluble βA plaques and neurofibrillary tangles (NFTs). Epidemiological data demonstrate that overlapping lesions diminish the βA plaque and NFT threshold necessary to precipitate clinical dementia. Moreover, a subset of persons who exhibit AD pathology remain resilient to disease while other persons with clinically-defined AD dementia do not exhibit AD-defining neuropathologic lesions. It is increasingly recognized that AD is a pathologically heterogeneous and biologically multifactorial disease with uncharacterized biologic phenomena involved in its genesis and progression. Here, we review the literature with regard to neuropathologic criteria and incipient AD changes, and discuss converging concepts regarding vascular and immune factors in AD.
Collapse
Affiliation(s)
- Rashi I. Mehta
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Rupal I. Mehta
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Raslan AA, Pham TX, Lee J, Hong J, Schmottlach J, Nicolas K, Dinc T, Bujor AM, Caporarello N, Thiriot A, von Andrian UH, Huang SK, Nicosia RF, Trojanowska M, Varelas X, Ligresti G. Single Cell Transcriptomics of Fibrotic Lungs Unveils Aging-associated Alterations in Endothelial and Epithelial Cell Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.523179. [PMID: 36712020 PMCID: PMC9882122 DOI: 10.1101/2023.01.17.523179] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Lung regeneration deteriorates with aging leading to increased susceptibility to pathologic conditions, including fibrosis. Here, we investigated bleomycin-induced lung injury responses in young and aged mice at single-cell resolution to gain insights into the cellular and molecular contributions of aging to fibrosis. Analysis of 52,542 cells in young (8 weeks) and aged (72 weeks) mice identified 15 cellular clusters, many of which exhibited distinct injury responses that associated with age. We identified Pdgfra + alveolar fibroblasts as a major source of collagen expression following bleomycin challenge, with those from aged lungs exhibiting a more persistent activation compared to young ones. We also observed age-associated transcriptional abnormalities affecting lung progenitor cells, including ATII pneumocytes and general capillary (gCap) endothelial cells (ECs). Transcriptional analysis combined with lineage tracing identified a sub-population of gCap ECs marked by the expression of Tropomyosin Receptor Kinase B (TrkB) that appeared in bleomycin-injured lungs and accumulated with aging. This newly emerged TrkB + EC population expressed common gCap EC markers but also exhibited a distinct gene expression signature associated with aberrant YAP/TAZ signaling, mitochondrial dysfunction, and hypoxia. Finally, we defined ACKR1 + venous ECs that exclusively emerged in injured lungs of aged animals and were closely associated with areas of collagen deposition and inflammation. Immunostaining and FACS analysis of human IPF lungs demonstrated that ACKR1 + venous ECs were dominant cells within the fibrotic regions and accumulated in areas of myofibroblast aggregation. Together, these data provide high-resolution insights into the impact of aging on lung cell adaptability to injury responses.
Collapse
|
16
|
Li L, Guo H, Lai B, Liang C, Chen H, Chen Y, Guo W, Yuan Z, Huang R, Zeng Z, Liang L, Zhao H, Zheng X, Li Y, Pu Q, Qi X, Cai D. Ablation of cardiomyocyte-derived BDNF during development causes myocardial degeneration and heart failure in the adult mouse heart. Front Cardiovasc Med 2022; 9:967463. [PMID: 36061561 PMCID: PMC9433718 DOI: 10.3389/fcvm.2022.967463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Brain-derived neurotrophic factor (BDNF) and its receptor TrkB-T1 were recently found to be expressed in cardiomyocytes. However, the functional role of cardiomyocyte-derived BDNF in heart pathophysiology is not yet fully known. Recent studies revealed that BDNF-TrkB pathway plays a critical role to maintain integrity of cardiac structure and function, cardiac pathology and regeneration of myocardial infarction (MI). Therefore, the BDNF-TrkB pathway may be a novel target for myocardial pathophysiology in the adult heart. Approach and results In the present study, we established a cardiomyocyte-derived BDNF conditional knockout mouse in which BDNF expression in developing cardiomyocytes is ablated under the control of the Myosin heavy chain 6 (MYH6) promoter. The results of the present study show that ablation of cardiomyocyte-derived BDNF during development does not impair survival, growth or reproduction; however, in the young adult heart, it causes cardiomyocyte death, degeneration of the myocardium, cardiomyocyte hypertrophy, left atrial appendage thrombosis, decreased cardiac function, increased cardiac inflammation and ROS activity, and metabolic disorders, leading to heart failure (HF) in the adult heart and eventually resulting in a decrease in the one-year survival rate. In addition, ablation of cardiomyocyte-derived BDNF during the developmental stage leads to exacerbation of cardiac dysfunction and poor regeneration after MI in adult hearts. Conclusion Cardiomyocyte-derived BDNF is irreplaceable for maintaining the integrity of cardiac structure and function in the adult heart and regeneration after MI. Therefore, the BDNF-TrkB pathway will be a novel target for myocardial pathophysiology in the adult heart.
Collapse
Affiliation(s)
- Lilin Li
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Hongyan Guo
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
- Jiangxi Provincial Key Laboratory of Medical Immunology and Immunotherapy, Jiangxi Academy of Medical Sciences, Nanchang, China
| | - Binglin Lai
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Chunbao Liang
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Hongyi Chen
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Yilin Chen
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Weimin Guo
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Ziqiang Yuan
- Department of Medical Oncology, Robert Wood Johnson of Medical School, Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Ruijin Huang
- Department of Neuroanatomy, Institute of Anatomy, University of Bonn, Bonn, Germany
- Department of Anatomy and Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Zhaohua Zeng
- Division of Cardiology, Department of Internal Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liying Liang
- Division of Cardiology, Department of Internal Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Zhao
- Stem Cell and Regeneration TRP, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xin Zheng
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Yanmei Li
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
| | - Qin Pu
- Department of Neuroanatomy, Institute of Anatomy, University of Bonn, Bonn, Germany
| | - Xufeng Qi
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
- *Correspondence: Xufeng Qi,
| | - Dongqing Cai
- Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
- Joint Laboratory for Regenerative Medicine, Chinese University of Hong Kong-Jinan University, Guangzhou, China
- International Base of Collaboration for Science and Technology (JNU), The Ministry of Science and Technology and Guangdong Province, Guangzhou, China
- Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, China
- Dongqing Cai,
| |
Collapse
|
17
|
Bazzari AH, Bazzari FH. BDNF Therapeutic Mechanisms in Neuropsychiatric Disorders. Int J Mol Sci 2022; 23:ijms23158417. [PMID: 35955546 PMCID: PMC9368938 DOI: 10.3390/ijms23158417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin in the adult brain and functions as both a primary neurotrophic signal and a neuromodulator. It serves essential roles in neuronal development, maintenance, transmission, and plasticity, thereby influencing aging, cognition, and behavior. Accumulating evidence associates reduced central and peripheral BDNF levels with various neuropsychiatric disorders, supporting its potential utilization as a biomarker of central pathologies. Subsequently, extensive research has been conducted to evaluate restoring, or otherwise augmenting, BDNF transmission as a potential therapeutic approach. Promising results were indeed observed for genetic BDNF upregulation or exogenous administration using a multitude of murine models of neurological and psychiatric diseases. However, varying mechanisms have been proposed to underlie the observed therapeutic effects, and many findings indicate the engagement of disease-specific and other non-specific mechanisms. This is because BDNF essentially affects all aspects of neuronal cellular function through tropomyosin receptor kinase B (TrkB) receptor signaling, the disruptions of which vary between brain regions across different pathologies leading to diversified consequences on cognition and behavior. Herein, we review the neurophysiology of BDNF transmission and signaling and classify the converging and diverging molecular mechanisms underlying its therapeutic potentials in neuropsychiatric disorders. These include neuroprotection, synaptic maintenance, immunomodulation, plasticity facilitation, secondary neuromodulation, and preservation of neurovascular unit integrity and cellular viability. Lastly, we discuss several findings suggesting BDNF as a common mediator of the therapeutic actions of centrally acting pharmacological agents used in the treatment of neurological and psychiatric illness.
Collapse
Affiliation(s)
- Amjad H. Bazzari
- Faculty of Medicine, Arab American University, 13 Zababdeh, Jenin 240, Palestine
- Correspondence:
| | - Firas H. Bazzari
- Faculty of Pharmacy, Arab American University, 13 Zababdeh, Jenin 240, Palestine;
| |
Collapse
|
18
|
Naletova I, Greco V, Sciuto S, Attanasio F, Rizzarelli E. Ionophore Ability of Carnosine and Its Trehalose Conjugate Assists Copper Signal in Triggering Brain-Derived Neurotrophic Factor and Vascular Endothelial Growth Factor Activation In Vitro. Int J Mol Sci 2021; 22:13504. [PMID: 34948299 PMCID: PMC8706131 DOI: 10.3390/ijms222413504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
l-carnosine (β-alanyl-l-histidine) (Car hereafter) is a natural dipeptide widely distributed in mammalian tissues and reaching high concentrations (0.7-2.0 mM) in the brain. The molecular features of the dipeptide underlie the antioxidant, anti-aggregating and metal chelating ability showed in a large number of physiological effects, while the biological mechanisms involved in the protective role found against several diseases cannot be explained on the basis of the above-mentioned properties alone, requiring further research efforts. It has been reported that l-carnosine increases the secretion and expression of various neurotrophic factors and affects copper homeostasis in nervous cells inducing Cu cellular uptake in keeping with the key metal-sensing system. Having in mind this l-carnosine ability, here we report the copper-binding and ionophore ability of l-carnosine to activate tyrosine kinase cascade pathways in PC12 cells and stimulate the expression of BDNF. Furthermore, the study was extended to verify the ability of the dipeptide to favor copper signaling inducing the expression of VEGF. Being aware that the potential protective action of l-carnosine is drastically hampered by its hydrolysis, we also report on the behavior of a conjugate of l-carnosine with trehalose that blocks the carnosinase degradative activity. Overall, our findings describe a copper tuning effect on the ability of l-carnosine and, particularly its conjugate, to activate tyrosine kinase cascade pathways.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research—CNR, Via Paolo Gaifami 18, 95126 Catania, Italy;
- National Inter-University Consortium Metals Chemistry in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126 Bari, Italy
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research—CNR, Via Paolo Gaifami 18, 95126 Catania, Italy;
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research—CNR, Via Paolo Gaifami 18, 95126 Catania, Italy;
- National Inter-University Consortium Metals Chemistry in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126 Bari, Italy
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| |
Collapse
|
19
|
Li X, Huang Y, Han Y, Yang Q, Zheng Y, Li W. LncPVT1 regulates osteogenic differentiation of human periodontal ligament cells via miR-10a-5p/brain-derived neurotrophic factor. J Periodontol 2021; 93:1093-1106. [PMID: 34793611 DOI: 10.1002/jper.21-0429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/02/2021] [Accepted: 11/07/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Identifying the factors affecting osteoblast differentiation ofperiodontal ligamentcells (PDLCs) can help enhance the regeneration of periodontal tissue.LncRNAplasmacytoma variant translocation 1 (lncPVT1) is an important regulatory factor involved in many biological processes, but its role in osteogenesisremains unclear. METHODS Expressionsof osteogenic markers were detected by quantitative reverse transcription polymerase chain reaction and Western blot analysis. Alkaline phosphatase staining was conducted for early osteoblast differentiation and alizarin red S staining was used for mineral deposition. RNA sequencing was used to identify the miRNAs regulated by lncPVT1 during osteogenesis. Cell transfection was used to overexpress or knockdown lncPVT1 and miR-10a-5p. Dual luciferase reporter assayswere conducted to analyze the binding of miR-10a-5p to brain-derived neurotrophic factor (BDNF). RESULTS LncPVT1 was significantly increased during osteogenic induction of PDLCs. Overexpression of lncPVT1 promoted osteogenesis, whereas lncPVT1 knockdown inhibited this process. RNA sequencing showed that miR-10a-5p expression was significantly increased after lncPVT1 knockdown.RNA immunoprecipitation assay further demonstrated the binding potential of lncPVT1 and miR-10a-5p. MiR-10a-5p inhibited the osteogenesis of PDLCs, and partially reversed the stimulatory effects of lncPVT1.Subsequently, we identified a predicted binding site for miR-10a-5p on BDNF and confirmed it using dual luciferase reporter assays. Moreover, lncPVT1 upregulated the expression of BDNF, while miR-10a-5p downregulated BDNF expression. BDNF promoted osteogenesis and partially rescued the si-lncPVT1-mediated inhibition of PDLCs osteogenic differentiation. CONCLUSION LncPVT1 positively regulated the osteogenic differentiation of PDLCs via miR-10a-5p and BDNF.Our resultsprovide a promising target for enhancing the osteogenic potential of PDLCs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xiaobei Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Yiping Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Yineng Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Qiaolin Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, P.R. China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, P.R. China
| |
Collapse
|
20
|
Sharma GP, Frei AC, Narayanan J, Gasperetti T, Veley D, Amjad A, Albano K, Fish BL, Himburg HA. Brain-derived neurotrophic factor promotes immune reconstitution following radiation injury via activation of bone marrow mesenchymal stem cells. PLoS One 2021; 16:e0259042. [PMID: 34695155 PMCID: PMC8544859 DOI: 10.1371/journal.pone.0259042] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/11/2021] [Indexed: 11/18/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the nerve growth factor family which has been extensively studied for its roles in neural development, long-term memory, brain injury, and neurodegenerative diseases. BDNF signaling through tropomyosin receptor kinase B (TrkB) stimulates neuronal cell survival. For this reason, small molecule TrkB agonists are under pre-clinical develoment for the treatment of a range of neurodegenerative diseases and injuries. Our laboratory recently reported BDNF is secreted by pro-regenerative endothelial progenitor cells (EPCs) which support hematopoietic reconstitution following total body irradiation (TBI). Here we report BDNF-TrkB signaling plays a novel regenerative role in bone marrow and thymic regeneration following radiation injury. Exogenous administration of BDNF or TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following myelosuppressive radiation injury promoted faster recovery of mature blood cells and hematopoietic stem cells capable of multi-lineage reconstitution. BDNF promotes hematopoietic regeneration via activation of PDGFRα+ bone marrow mesenchymal stem cells (MSCs) which increase secretion of hematopoietic cytokines interleukin 6 (IL-6) and leukemia inhibitory factor (LIF) in response to TrkB activation. These data suggest pharmacologic activation of the BDNF pathway with either BDNF or 7,8-DHF may be beneficial for treatment of radiation or chemotherapy induced myelosuppression.
Collapse
Affiliation(s)
- Guru Prasad Sharma
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Anne C. Frei
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Jayashree Narayanan
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Tracy Gasperetti
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Dana Veley
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Asma Amjad
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Katherine Albano
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Brian L. Fish
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Heather A. Himburg
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
21
|
The brain-derived neurotrophic factor prompts platelet aggregation and secretion. Blood Adv 2021; 5:3568-3580. [PMID: 34546355 DOI: 10.1182/bloodadvances.2020004098] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has both autocrine and paracrine roles in neurons, and its release and signaling mechanisms have been extensively studied in the central nervous system. Large quantities of BDNF have been reported in circulation, essentially stored in platelets with concentrations reaching 100- to 1000-fold those of neurons. Despite this abundance, the function of BDNF in platelet biology has not been explored. At low concentrations, BDNF primed platelets, acting synergistically with classical agonists. At high concentrations, BDNF induced complete biphasic platelet aggregation that in part relied on amplification from secondary mediators. Neurotrophin-4, but not nerve growth factor, and an activating antibody against the canonical BDNF receptor tropomyosin-related kinase B (TrkB) induced similar platelet responses to BDNF, suggesting TrkB could be the mediator. Platelets expressed, both at their surface and in their intracellular compartment, a truncated form of TrkB lacking its tyrosine kinase domain. BDNF-induced platelet aggregation was prevented by inhibitors of Ras-related C3 botulinum toxin substrate 1 (Rac1), protein kinase C, and phosphoinositide 3-kinase. BDNF-stimulated platelets secreted a panel of angiogenic and inflammatory cytokines, which may play a role in maintaining vascular homeostasis. Two families with autism spectrum disorder were found to carry rare missense variants in the BDNF gene. Platelet studies revealed defects in platelet aggregation to low concentrations of collagen, as well as reduced adenosine triphosphate secretion in response to adenosine diphosphate. In summary, circulating BDNF levels appear to regulate platelet activation, aggregation, and secretion through activation of a truncated TrkB receptor and downstream kinase-dependent signaling.
Collapse
|
22
|
Abstract
Exosomes are nanoscale extracellular vesicles that can transport cargos of proteins, lipids, DNA, various RNA species and microRNAs (miRNAs). Exosomes can enter cells and deliver their contents to recipient cell. Owing to their cargo exosomes can transfer different molecules to the target cells and change the phenotype of these cells. The fate of the contents of an exosome depends on its target destination. Various mechanisms for exosome uptake by target cells have been proposed, but the mechanisms responsible for exosomes internalization into cells are still debated. Exosomes exposed cells produce labeled protein kinases, which are expressed by other cells. This means that these kinases are internalized by exosomes, and transported into the cytoplasm of recipient cells. Many studies have confirmed that exosomes are not only secreted by living cells, but also internalized or accumulated by the other cells. The "next cell hypothesis" supports the notion that exosomes constitute communication vehicles between neighboring cells. By this mechanism, exosomes participate in the development of diabetes and its associated complications, critically contribute to the spreading of neuronal damage in Alzheimer's disease, and non-proteolysed form of Fas ligand (mFasL)-bearing exosomes trigger the apoptosis of T lymphocytes. Furthermore, exosomes derived from human B lymphocytes induce antigen-specific major histocompatibility complex (MHC) class II-restricted T cell responses. Interestingly, exosomes secreted by cancer cells have been demonstrated to express tumor antigens, as well as immune suppressive molecules. This process is defined as "exosome-immune suppression" concept. The interplay via the exchange of exosomes between cancer cells and between cancer cells and the tumor stroma promote the transfer of oncogenes and onco-miRNAs from one cell to other. Circulating exosomes that are released from hypertrophic adipocytes are effective in obesity-related complications. On the other hand, the "inflammasome-induced" exosomes can activate inflammatory responses in recipient cells. In this chapter protein kinases-related checkpoints are emphasized considering the regulation of exosome biogenesis, secretory traffic, and their impacts on cell death, tumor growth, immune system, and obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey.
| |
Collapse
|
23
|
Pius-Sadowska E, Machaliński B. Pleiotropic activity of nerve growth factor in regulating cardiac functions and counteracting pathogenesis. ESC Heart Fail 2021; 8:974-987. [PMID: 33465292 PMCID: PMC8006610 DOI: 10.1002/ehf2.13138] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 11/05/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022] Open
Abstract
Cardiac innervation density generally reflects the levels of nerve growth factor (NGF) produced by the heart—changes in NGF expression within the heart and vasculature contribute to neuronal remodelling (e.g. sympathetic hyperinnervation or denervation). Its synthesis and release are altered under different pathological conditions. Although NGF is well known for its survival effects on neurons, it is clear that these effects are more wide ranging. Recent studies reported both in vitro and in vivo evidence for beneficial actions of NGF on cardiomyocytes in normal and pathological hearts, including prosurvival and antiapoptotic effects. NGF also plays an important role in the crosstalk between the nervous and cardiovascular systems. It was the first neurotrophin to be implicated in postnatal angiogenesis and vasculogenesis by autocrine and paracrine mechanisms. In connection with these unique cardiovascular properties of NGF, we have provided comprehensive insight into its function and potential effect of NGF underlying heart sustainable/failure conditions. This review aims to summarize the recent data on the effects of NGF on various cardiovascular neuronal and non‐neuronal functions. Understanding these mechanisms with respect to the diversity of NGF functions may be crucial for developing novel therapeutic strategies, including NGF action mechanism‐guided therapies.
Collapse
Affiliation(s)
- Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, Szczecin, 70111, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, Szczecin, 70111, Poland
| |
Collapse
|
24
|
Trombetta IC, DeMoura JR, Alves CR, Carbonari-Brito R, Cepeda FX, Lemos JR. Serum Levels of BDNF in Cardiovascular Protection and in Response to Exercise. Arq Bras Cardiol 2020; 115:263-269. [PMID: 32876194 PMCID: PMC8384297 DOI: 10.36660/abc.20190368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 03/16/2020] [Indexed: 12/17/2022] Open
Abstract
As doenças cardiovasculares (DCV) são atualmente a maior causa de morte no Brasil e no mundo. Em 2016 as DCV foram responsáveis por mais de 17 milhões de mortes, representando 31% de todas as mortes em nível global. Mecanismos moleculares e genéticos podem estar envolvidos na proteção cardiovascular e devem ser considerados nas novas abordagens terapêuticas. Nesse sentido, recentes estudos têm relatado que o Fator Neurotrófico Derivado do Encéfalo (Brain-Derived Neurotrophic Factor, BDNF) está reduzido em indivíduos predispostos a desenvolverem DCV, e que o treinamento físico aeróbio aumenta as quantidades de BDNF circulante. O BDNF é uma neurotrofina encontrada em altas concentrações no hipocampo e córtex cerebral, sendo considerada molécula-chave na manutenção da plasticidade sináptica e na sobrevivência das células neuronais. Além da plasticidade neuronal, BDNF também é importante na função vascular, promovendo angiogênese por meio da regulação por espécies reativas de oxigênio (ROS). Entretanto, uma variante do gene do BDNF em humanos, o polimorfismo Val66Met (substituição do aminoácido valina por uma metionina na posição 66 do códon), que ocorre em 20-30% da população caucasiana, pode afetar as concentrações de BDNF no plasma e sua atividade em todos os tecidos periféricos contendo receptores tirosina quinase B (TrkB), como o endotélio. De fato, recentemente observamos que o polimorfismo Val66Met prejudica a reatividade vascular e o BDNF circulante em resposta ao treinamento físico. Dessa forma, apresentaremos a seguir uma discussão sobre os níveis séricos de BDNF na proteção cardiovascular, a variante genética Val66Met na reatividade vascular e o efeito do exercício físico.
Collapse
Affiliation(s)
| | - José Roberto DeMoura
- Universidade Nove de Julho (UNINOVE), São Paulo, SP - Brasil.,Escola de Educação Física da Polícia Militar do Estado de São Paulo, São Paulo, SP - Brasil
| | | | | | | | - José Ribeiro Lemos
- Escola de Educação Física da Polícia Militar do Estado de São Paulo, São Paulo, SP - Brasil
| |
Collapse
|
25
|
Herland A, Maoz BM, FitzGerald EA, Grevesse T, Vidoudez C, Sheehy SP, Budnik N, Dauth S, Mannix R, Budnik B, Parker KK, Ingber DE. Proteomic and Metabolomic Characterization of Human Neurovascular Unit Cells in Response to Methamphetamine. ACTA ACUST UNITED AC 2020; 4:e1900230. [PMID: 32744807 DOI: 10.1002/adbi.201900230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 07/02/2020] [Indexed: 01/31/2023]
Abstract
The functional state of the neurovascular unit (NVU), composed of the blood-brain barrier and the perivasculature that forms a dynamic interface between the blood and the central nervous system (CNS), plays a central role in the control of brain homeostasis and is strongly affected by CNS drugs. Human primary brain microvascular endothelium, astrocyte, pericyte, and neural cell cultures are often used to study NVU barrier functions as well as drug transport and efficacy; however, the proteomic and metabolomic responses of these different cell types are not well characterized. Culturing each cell type separately, using deep coverage proteomic analysis and characterization of the secreted metabolome, as well as measurements of mitochondrial activity, the responses of these cells under baseline conditions and when exposed to the NVU-impairing stimulant methamphetamine (Meth) are analyzed. These studies define the previously unknown metabolic and proteomic profiles of human brain pericytes and lead to improved characterization of the phenotype of each of the NVU cell types as well as cell-specific metabolic and proteomic responses to Meth.
Collapse
Affiliation(s)
- Anna Herland
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden.,AIMES, Center for the Advancement of Integrated Engineering and Medical Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, 17177, Sweden
| | - Ben M Maoz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.,Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel.,Department of Biomedical Engineering, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Edward A FitzGerald
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Thomas Grevesse
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Charles Vidoudez
- Small Molecule Mass Spectrometry Facility, Harvard University, Cambridge, MA, 02138, USA
| | - Sean P Sheehy
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Nikita Budnik
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Stephanie Dauth
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Robert Mannix
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Bogdan Budnik
- Mass Spectrometry and Proteomics Resource Laboratory, Harvard University, Cambridge, MA, 02138, USA
| | - Kevin Kit Parker
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Vascular Biology Program and Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
26
|
Antioxidant properties of plant polyphenols in the counteraction of alcohol-abuse induced damage: Impact on the Mediterranean diet. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
27
|
Bonafina A, Paratcha G, Ledda F. Deciphering New Players in the Neurogenic Adult Hippocampal Niche. Front Cell Dev Biol 2020; 8:548. [PMID: 32714932 PMCID: PMC7346873 DOI: 10.3389/fcell.2020.00548] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/10/2020] [Indexed: 12/23/2022] Open
Abstract
In the mammalian adult hippocampus, new neurons are continuously generated throughout life in the subgranular zone of the dentate gyrus. Increasing evidence point out the contribution of adult-born hippocampal granule cells (GCs) to cognitive processes such as learning and memory, indicating the relevance of understanding the molecular mechanisms that control the development of these new neurons in the preexisting hippocampal circuits. Cell proliferation and functional integration of adult-born GCs is a process highly regulated by different intrinsic and extrinsic factors. In this review, we discuss recent advances related with cellular components and extrinsic signals of the hippocampal neurogenic niche that support and modulate neurogenesis under physiological conditions.
Collapse
Affiliation(s)
- Antonela Bonafina
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Gustavo Paratcha
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Fernanda Ledda
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
28
|
Prognostic significance of brain-derived neurotrophic factor levels in patients with heart failure and reduced left ventricular ejection fraction. Anatol J Cardiol 2020; 22:309-316. [PMID: 31789613 PMCID: PMC6955046 DOI: 10.14744/anatoljcardiol.2019.37941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective: Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family. The aim of the present study was to investigate the relationship between BDNF levels and prognostic markers in patients with heart failure (HF) and reduced left ventricular ejection fraction (LVEF), considering death or rehospitalization due to HF. Methods: Patients with severe left ventricular systolic dysfunction (LVEF ≤35%) and individuals with no history of cardiac disease (control group) were included in the study conducted between 2013 and 2017. Of the included patients, 52 were classified as mildly symptomatic [New York Heart Association (NYHA) I–II], and 108 were classified as severely symptomatic (NYHA III). The control group comprised 50 individuals. The primary endpoints of the study consisted of cardiovascular death during long-term follow-up and hospitalization for worsening of HF. Results: The mean age of the patient group was 67.60±11.45 years and 58% were male, whereas that of the control group was 66.28±11.30 years and 48% were male. The N-terminal pro-brain-type natriuretic peptide (NT-pro-BNP) serum levels in patients with HF were higher, whereas the BDNF values were lower than those in the control group (NT-pro-BNP: 5010±851 pg/mL vs. 33±11 pg/mL, p<0.001; BDNF: 8.64±1.12 ng/mL vs. 17.58±4.51 ng/mL, p<0.001). Multivariable analysis suggested that there was a significant association between BDNF levels and clinical status, generating the primary endpoints of death [BDNF levels: Odds ratio (OR)=0.17, 95% confidence interval (CI): 0.05–0.53, p=0.002], and rehospitalization (BDNF levels: OR=0.702, 95%CI: 0.54–0.92, p=0.010). Conclusion: Decreased serum BDNF levels were associated with death and rehospitalization in patients with HF, suggesting that these levels can be useful prognostic biomarkers.
Collapse
|
29
|
D'Angelo A, Ceccanti M, Petrella C, Greco A, Tirassa P, Rosso P, Ralli M, Ferraguti G, Fiore M, Messina MP. Role of neurotrophins in pregnancy, delivery and postpartum. Eur J Obstet Gynecol Reprod Biol 2020; 247:32-41. [PMID: 32058187 DOI: 10.1016/j.ejogrb.2020.01.046] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 01/09/2023]
Abstract
Neurotrophins (NTs) are a family of polypeptides whose functions have been extensively studied in the past two decades. In particular, Nerve Growth Factor (NGF) and Brain-Derived Neurotrophic Factor (BDNF) play a major role in the development, nutrition and growth of the central and peripheral nervous system and in the pathogenesis of neurodegenerative, cardiometabolic and (auto)immune diseases. However, NGF and BDNF have subtle functions for follicular development, implantation, and placentation. This short narrative review summarizes the existing evidence, published between 2000 and 2019, about the role of NTs in many different conditions that might affect women during and after pregnancy such as preeclampsia, gestational diabetes, obesity, depression, anxiety, smoking and alcohol abuse. Literature suggests that the dysregulation of synthesis and release of NTs may lead to decisive effects on both maternal and fetal health. Some piece of evidences was found about a possible association between NGF/BDNF and breastfeeding. Additional studies on human models are necessary to further characterize the role of NTs in life-changing experiences like labor and delivery.
Collapse
Affiliation(s)
- Alessio D'Angelo
- Department of Gynecology, Obstetric, and Urology, Sapienza University of Rome, Italy
| | - Mauro Ceccanti
- Centro Riferimento Alcologico Regione Lazio, Sapienza University of Rome, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Paola Tirassa
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| | - Pamela Rosso
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Italy
| | | | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy.
| | | |
Collapse
|
30
|
Liu H, Tan N, Xu D, Li CY, Xian GJ. NGF and CNTF expression and regulation mechanism by miRNA in acute paralytic strabismus. Int Ophthalmol 2020; 40:975-984. [PMID: 31925656 DOI: 10.1007/s10792-019-01270-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/30/2019] [Indexed: 11/24/2022]
Abstract
BACKGROUND Nerve growth factor (NGF) and ciliary neurotrophic factor (CNTF) are well-known neurotrophic factors and widely used in the clinical treatment for its promotion effect on peripheral nerve regeneration. And they were also recommended for the acute paralytic strabismus treatment. However, whether the NGF and CNTF have protective effect for the extraocular muscles of acute paralytic strabismus patients is still poorly understood. PURPOSE In this study, we want to evaluate the biological function of NGF and CNTF on the extraocular muscle cells and reveale the regulation mechanism behind it. METHODS Firstly, the relative expression of ngf and cntf was assessed by quantitative real-time RT-PCR. Then, the influence of NGF and CNTF on the extraocular muscle cell proliferation was determined by CCK-8. The inflammatory response in muscle cells after NGF and CNTF treatment was evaluated by ELISA and ROS detection. In addition to this, the up-stream regulation of the ngf and cntf expression was also studied. The TargetScan was used for the predication of potential miRNAs targeting with ngf and cntf 30-UTR, which is soon confirmed by luciferase activity assay. RESULTS all the results in this research indicated that NGF and CNTF could promote the muscle cell proliferation and inhibit the inflammatory levels, then exert protective effect on the muscle cell function. RESULTS All the results in this research indicated that NGF and CNTF could promote the muscle cell proliferation and inhibit the inflammatory levels, then exert protective effect on the muscle cell function. CONCLUSION It was conceivable that let 7-5p was the up-stream regulator of ngf and cntf, and let 7-5p might serve as a potential molecular target for acute paralytic strabismus treatment.
Collapse
Affiliation(s)
- Hua Liu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| | - Nian Tan
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| | - Duo Xu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China.
| | - Chong-Yi Li
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| | - Guang-Jun Xian
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| |
Collapse
|
31
|
Dangat K, Khaire A, Joshi S. Cross talk of vascular endothelial growth factor and neurotrophins in mammary gland development. Growth Factors 2020; 38:16-24. [PMID: 32646254 DOI: 10.1080/08977194.2020.1792469] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During the period of lactation, there is extensive growth and development of the mammary gland in order to fulfil the increased demands of milk for the growing infant. Angiogenesis plays a key role in alveolar development and facilitates optimal milk production. Vascular endothelial growth factor (VEGF) is one of the key growth factors regulating angiogenesis in mammary gland. Apart from VEGF, neurotrophins are also known to regulate angiogenesis through direct or indirect mechanisms. Few studies have demonstrated mRNA levels of neurotrophins and their receptors in mammary gland both in humans and rodents. A cross talk between VEGF and neurotrophins has been described in placental development. The enteric and central nervous system are not fully developed at birth, making it imperative to have appropriate levels of angiogenic factors and neurotrophins during postnatal period. The current review summarises studies which describe the role of neurotrophins and angiogenic factors in the mammary gland development.
Collapse
Affiliation(s)
- Kamini Dangat
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune, India
| | - Amrita Khaire
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune, India
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune, India
| |
Collapse
|
32
|
Neurons can upregulate Cav-1 to increase intake of endothelial cells-derived extracellular vesicles that attenuate apoptosis via miR-1290. Cell Death Dis 2019; 10:869. [PMID: 31740664 PMCID: PMC6861259 DOI: 10.1038/s41419-019-2100-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/02/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) including exosomes can serve as mediators of cell–cell communication under physiological and pathological conditions. However, cargo molecules carried by EVs to exert their functions, as well as mechanisms for their regulated release and intake, have been poorly understood. In this study, we examined the effects of endothelial cells-derived EVs on neurons suffering from oxygen-glucose deprivation (OGD), which mimics neuronal ischemia-reperfusion injury in human diseases. In a human umbilical endothelial cell (HUVEC)–neuron coculture assay, we found that HUVECs reduced apoptosis of neurons under OGD, and this effect was compromised by GW4869, a blocker of exosome release. Purified EVs could be internalized by neurons and alleviate neuronal apoptosis under OGD. A miRNA, miR-1290, was highly enriched in HUVECs-derived EVs and was responsible for EV-mediated neuronal protection under OGD. Interestingly, we found that OGD enhanced intake of EVs by neurons cultured in vitro. We examined the expression of several potential receptors for EV intake and found that caveolin-1 (Cav-1) was upregulated in OGD-treated neurons and mice suffering from middle cerebral artery occlusion (MCAO). Knock-down of Cav-1 in neurons reduced EV intake, and canceled EV-mediated neuronal protection under OGD. HUVEC-derived EVs alleviated MCAO-induced neuronal apoptosis in vivo. These findings suggested that ischemia likely upregulates Cav-1 expression in neurons to increase EV intake, which protects neurons by attenuating apoptosis via miR-1290.
Collapse
|
33
|
Ramezani M, Emami Zeydi A. Brain-Derived Neurotrophic Factor: A Promising Biomarker for Predicting Prematurity-Related Complications. Int J Prev Med 2019; 10:140. [PMID: 31516681 PMCID: PMC6710922 DOI: 10.4103/ijpvm.ijpvm_421_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 04/09/2019] [Indexed: 11/06/2022] Open
Affiliation(s)
- Monir Ramezani
- Nursing and Midwifery Care Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pediatric Nursing, School of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Emami Zeydi
- Department of Medical-Surgical Nursing, Nasibeh School of Nursing and Midwifery, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
34
|
Wang X, Prager BC, Wu Q, Kim LJY, Gimple RC, Shi Y, Yang K, Morton AR, Zhou W, Zhu Z, Obara EAA, Miller TE, Song A, Lai S, Hubert CG, Jin X, Huang Z, Fang X, Dixit D, Tao W, Zhai K, Chen C, Dong Z, Zhang G, Dombrowski SM, Hamerlik P, Mack SC, Bao S, Rich JN. Reciprocal Signaling between Glioblastoma Stem Cells and Differentiated Tumor Cells Promotes Malignant Progression. Cell Stem Cell 2019; 22:514-528.e5. [PMID: 29625067 DOI: 10.1016/j.stem.2018.03.011] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 01/19/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Glioblastoma is the most lethal primary brain tumor; however, the crosstalk between glioblastoma stem cells (GSCs) and their supportive niche is not well understood. Here, we interrogated reciprocal signaling between GSCs and their differentiated glioblastoma cell (DGC) progeny. We found that DGCs accelerated GSC tumor growth. DGCs preferentially expressed brain-derived neurotrophic factor (BDNF), whereas GSCs expressed the BDNF receptor NTRK2. Forced BDNF expression in DGCs augmented GSC tumor growth. To determine molecular mediators of BDNF-NTRK2 paracrine signaling, we leveraged transcriptional and epigenetic profiles of matched GSCs and DGCs, revealing preferential VGF expression by GSCs, which patient-derived tumor models confirmed. VGF serves a dual role in the glioblastoma hierarchy by promoting GSC survival and stemness in vitro and in vivo while also supporting DGC survival and inducing DGC secretion of BDNF. Collectively, these data demonstrate that differentiated glioblastoma cells cooperate with stem-like tumor cells through BDNF-NTRK2-VGF paracrine signaling to promote tumor growth.
Collapse
Affiliation(s)
- Xiuxing Wang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Briana C Prager
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Qiulian Wu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Leo J Y Kim
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, and The Key Laboratory of Tumor Immunopathology, The Ministry of Education of China, Chongqing, China
| | - Kailin Yang
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Andrew R Morton
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Wenchao Zhou
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Zhe Zhu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | | | - Tyler E Miller
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Anne Song
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Sisi Lai
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Christopher G Hubert
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Xun Jin
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Zhi Huang
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Xiaoguang Fang
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Deobrat Dixit
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Weiwei Tao
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Kui Zhai
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Cong Chen
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Zhen Dong
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Guoxin Zhang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Stephen M Dombrowski
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Petra Hamerlik
- Brain Tumor Biology, Danish Cancer Society Research Center, Strandboulevarden 49, Copenhagen 2100, Denmark
| | - Stephen C Mack
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Shideng Bao
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
35
|
Segatto M, Fico E, Gharbiya M, Rosso P, Carito V, Tirassa P, Plateroti R, Lambiase A. VEGF inhibition alters neurotrophin signalling pathways and induces caspase-3 activation and autophagy in rabbit retina. J Cell Physiol 2019; 234:18297-18307. [PMID: 30891770 DOI: 10.1002/jcp.28462] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/26/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
This study sought to evaluate the prospective role exerted by vascular endothelial growth factor (VEGF) in the modulation of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) signalling pathways in the rabbit retina. To reach this aim, the anti-VEGF agents aflibercept and ranibizumab were used as a pharmacological approach to evaluate the putative consequences elicited by VEGF inhibition on neurotrophin signalling. VEGF inhibition determined a marked imbalance in proneurotrophin expression, a significant reduction in TrkA and TrkB phosphorylation states and a decrease in the pan-neurotrophin receptor p75. Importantly, VEGF blockade also caused a strong increase in cleaved caspase-3, beclin-1 and lipidated LC3. The effects were more pronounced in the aflibercept group when compared with ranibizumab-treated rabbits, particularly 1 week after injection. This study demonstrates that VEGF exerts pivotal physiological roles in regulating NGF and BDNF pathways in the retina, as its inhibition by anti-VEGF agents deeply impacts neurotrophin homeostasis. These events are accompanied by a sustained induction of apoptotic and autophagic markers, suggesting that anti-VEGF-dependent impairments in neurotrophin signalling could be responsible for the activation of retinal cell death pathways.
Collapse
Affiliation(s)
- Marco Segatto
- Department of Biosciences and Territory, University of Molise, Pesche, Italy.,Department of Sense Organs, University of Rome "La Sapienza", Rome, Italy
| | - Elena Fico
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Magda Gharbiya
- Department of Sense Organs, University of Rome "La Sapienza", Rome, Italy
| | - Pamela Rosso
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy
| | - Valentina Carito
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy
| | - Paola Tirassa
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy
| | - Rocco Plateroti
- Department of Sense Organs, University of Rome "La Sapienza", Rome, Italy
| | | |
Collapse
|
36
|
Mehta D, Granstein RD. Immunoregulatory Effects of Neuropeptides on Endothelial Cells: Relevance to Dermatological Disorders. Dermatology 2019; 235:175-186. [PMID: 30808842 DOI: 10.1159/000496538] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/31/2018] [Indexed: 11/19/2022] Open
Abstract
Many skin diseases, including psoriasis and atopic dermatitis, have a neurogenic component. In this regard, bidirectional interactions between components of the nervous system and multiple target cells in the skin and elsewhere have been receiving increasing attention. Neuropeptides released by sensory nerves that innervate the skin can directly modulate functions of keratinocytes, Langerhans cells, dermal dendritic cells, mast cells, dermal microvascular endothelial cells and infiltrating immune cells. As a result, neuropeptides and neuropeptide receptors participate in a complex, interdependent network of mediators that modulate the skin immune system, skin inflammation, and wound healing. In this review, we will focus on recent studies demonstrating the roles of α-melanocyte-stimulating hormone, calcitonin gene-related peptide, substance P, somatostatin, vasoactive intestinal peptide, pituitary adenylate cyclase-activating peptide, and nerve growth factor in modulating inflammation and immunity in the skin through their effects on dermal microvascular endothelial cells.
Collapse
Affiliation(s)
- Devina Mehta
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | | |
Collapse
|
37
|
Marie C, Pedard M, Quirié A, Tessier A, Garnier P, Totoson P, Demougeot C. Brain-derived neurotrophic factor secreted by the cerebral endothelium: A new actor of brain function? J Cereb Blood Flow Metab 2018; 38:935-949. [PMID: 29557702 PMCID: PMC5998997 DOI: 10.1177/0271678x18766772] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Low cerebral levels of brain-derived neurotrophic factor (BDNF), which plays a critical role in many brain functions, have been implicated in neurodegenerative, neurological and psychiatric diseases. Thus, increasing BDNF levels in the brain is considered an attractive possibility for the prevention/treatment of various brain diseases. To date, BDNF-based therapies have largely focused on neurons. However, given the cross-talk between endothelial cells and neurons and recent evidence that BDNF expressed by the cerebral endothelium largely accounts for BDNF levels present in the brain, it is likely that BDNF-based therapies would be most effective if they also targeted the cerebral endothelium. In this review, we summarize the available knowledge about the biology and actions of BDNF derived from endothelial cells of the cerebral microvasculature and we emphasize the remaining gaps and shortcomings.
Collapse
Affiliation(s)
- Christine Marie
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France
| | - Martin Pedard
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France.,2 Service de Neurologie, CHRU, Dijon, France
| | - Aurore Quirié
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France
| | - Anne Tessier
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France
| | | | - Perle Totoson
- 3 EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
| | - Céline Demougeot
- 3 EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
38
|
Pedard M, Quirié A, Totoson P, Verhoeven F, Garnier P, Tessier A, Demougeot C, Marie C. Vascular brain-derived neurotrophic factor pathway in rats with adjuvant-induced arthritis: Effect of anti-rheumatic drugs. Atherosclerosis 2018; 274:77-85. [PMID: 29753231 DOI: 10.1016/j.atherosclerosis.2018.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/11/2018] [Accepted: 05/01/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS In rheumatoid arthritis, the control of both disease activity and standard cardiovascular (CV) risk factors is expected to attenuate the increased CV risk. Evidence that brain-derived neurotrophic factor (BDNF) plays a role in vascular biology led us to investigate the vascular BDNF pathway in arthritis rats as well as the interaction between endothelial nitric oxide (NO) and BDNF production. METHODS The aortic BDNF pathway was studied in rats with adjuvant-induced arthritis, (AIA) using Western blot and immunohistochemical analysis. Control of arthritis score was achieved by administration (for 3 weeks) of an equipotent dosage of etanercept, prednisolone, methotrexate, celecoxib or diclofenac. Aortas were exposed to an NO donor or an NO synthase inhibitor and vasoreactivity experiments were performed using LM22A-4 as a TrkB agonist. RESULTS Vascular BDNF and full length tropomyosin-related kinase B receptor (TrkB-FL) were higher in AIA than in control rats. These changes coincided with decreased endothelial immunoreactivity in BDNF and pTrkBtyr816 and were disconnected from arthritis score. Among anti-rheumatic drugs, only prednisolone and methotrexate prevented AIA-induced vascular BDNF loss. The effect of AIA on aortic BDNF levels was reversed by an NO donor and reproduced by an NOS inhibitor. Finally, LM22A-4 induced both NO-dependent vasodilation and phosphorylation of endothelial NO synthase at serine 1177. CONCLUSIONS Our study identified changes in the BDNF/TrkB pathway as a disease activity-independent component of AIA-associated changes in endothelial phenotype. It provides new perspectives in the understanding and management of the high CV risk reported in rheumatoid arthritis.
Collapse
Affiliation(s)
- Martin Pedard
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000, Dijon, France; Service de Neurologie, CHRU, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000, Dijon, France
| | - Perle Totoson
- EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France
| | - Frank Verhoeven
- EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000, Dijon, France
| | - Anne Tessier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000, Dijon, France
| | - Céline Demougeot
- EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030, Besançon, France
| | - Christine Marie
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, F-21000, Dijon, France.
| |
Collapse
|
39
|
Hypoxia and Local Inflammation in Pulmonary Artery Structure and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:325-334. [PMID: 29047096 DOI: 10.1007/978-3-319-63245-2_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxia is recognized as a contributor to pulmonary vascular diseases such as pulmonary hypertension. Hypoxia-induced inflammatory changes can enhance structural and functional changes in pulmonary artery (PA) in the context of PH. Accordingly, understanding how hypoxia and inflammation are linked in the context of pulmonary artery structure and function could be relevant towards development of novel therapies for PH. In this regard, factors such as thymic stromal lymphopoietin (TSLP), an inflammatory cytokine, and brain-derived neurotrophic factor (BDNF), a neurotrophin, have been found critical for nonvascular systems such as airway and asthma. While TSLP canonically affects the immune system, in nonvascular systems, noncanonical effects such as altered [Ca2+]i and cell proliferation have been noted: aspects also relevant to the PA, where there is currently little to no data. Similarly, better known in the nervous system, there is increasing evidence that BDNF is locally produced by structural cells of the airway and can contribute to asthma pathophysiology. In this chapter, we summarize the potential relevance of factors such as TSLP and BDNF to the PA and in the context of hypoxia influences towards development of PH. We focus on cell sources and targets such as PA endothelial cells (PAECs) and smooth muscle cells (PASMCs), and the effects of TSLP or BDNF on intracellular Ca2+ responses to vasoconstrictor agonist, cell proliferation, and potential signaling cascades involved.
Collapse
|
40
|
Becerra-Calixto A, Posada-Duque R, Cardona-Gómez GP. Recovery of Neurovascular Unit Integrity by CDK5-KD Astrocyte Transplantation in a Global Cerebral Ischemia Model. Mol Neurobiol 2018; 55:8563-8585. [PMID: 29564811 DOI: 10.1007/s12035-018-0992-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/06/2018] [Indexed: 12/16/2022]
Abstract
Astrocytes play metabolic and structural support roles and contribute to the integrity of the blood-brain barrier (BBB), linking communication between neurons and the endothelium. Cyclin-dependent kinase 5 (CDK5) likely exerts a dual effect on the endothelium and astrocytes due to its involvement in migration and angiogenesis; the overactivation of CDK5 is associated with dysfunction in glutamate recapture and hypoxia. Recently, we proposed that CDK5-targeted astrocytes facilitate the recovery of neurological and motor function in transplanted ischemic rats. In the current study, we treated cerebral ischemic rats and endothelial cells exposed to glutamate toxicity with CDK5 knock-down (CDK5-KD) astrocytes to determine the role of CDK5 in neurovascular integrity. We found that the effects of CDK5-KD were sustained for 4 months, preventing neuronal and astrocyte loss, facilitating the recovery of the BBB via the production of BDNF by endogenous astrocytes (GFP-) surrounding vessels in the motor cortex and the corpus callosum of global ischemic rats, and improving neurological performance. These findings were supported by the in vitro findings of increased transendothelial resistance, p120-ctn+ adhesion and reduced intercellular gaps induced by a CDK5 inhibitor (roscovitine) in bEnd.3 cells in a glutamate-toxicity model. Additionally, CDK5-KD astrocytes in co-culture protected the endothelial cell viability, increased BDNF release from astrocytes, increased BDNF immunoreactivity in neighboring astrocytes and endothelial cells and enhanced cell adhesion in a glutamate-toxicity model. Altogether, these findings suggest that a CDK5 reduction in astrocytes protects the endothelium, which promotes BDNF release, endothelial adhesion, and the recovery of neurovascular unit integrity and brain function in ischemic rats.
Collapse
Affiliation(s)
- Andrea Becerra-Calixto
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, SIU, University of Antioquia, Calle 70, No. 52-21, Medellin, Colombia
| | - Rafael Posada-Duque
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, SIU, University of Antioquia, Calle 70, No. 52-21, Medellin, Colombia.,Institute of Biology, Faculty of Exact and Natural Sciences, University of Antioquia, Medellin, Colombia
| | - Gloria Patricia Cardona-Gómez
- Neuroscience Group of Antioquia, Cellular and Molecular Neurobiology Area, Faculty of Medicine, SIU, University of Antioquia, Calle 70, No. 52-21, Medellin, Colombia. .,Universidad de Antioquia, Sede de Investigación Universitaria (SIU), Calle 62 # 52 - 59; Torre 1, Piso 4, Laboratorio 412, Medellín, Colombia.
| |
Collapse
|
41
|
Mori A, Nishioka Y, Yamada M, Nishibata Y, Masuda S, Tomaru U, Honma N, Moriyama T, Ishizu A. Brain-derived neurotrophic factor induces angiogenin secretion and nuclear translocation in human umbilical vein endothelial cells. Pathol Res Pract 2018; 214:521-526. [PMID: 29573867 DOI: 10.1016/j.prp.2018.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/25/2018] [Accepted: 02/14/2018] [Indexed: 11/17/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a well-known humoral protein that induces growth of neurons. Recent studies have suggested that BDNF could act as an angiogenesis inducer similar to vascular endothelial growth factor (VEGF). Angiogenin is a strong mediator of angiogenesis. It has particular characteristics both as a secreted protein and a transcription factor. After being incorporated into the cytoplasm, angiogenin is immediately transferred to the nucleus and then mediates the angiogenic effects of angiogenesis inducers, including VEGF. The aim of this study is to determine the association between BDNF and angiogenin. At first, we determined the secretion of angiogenin from human umbilical vein endothelial cells (HUVEC) induced by BDNF with enzyme-linked immunosorbent assay. Next, we determined BDNF-induced nuclear translocation of angiogenin by immunofluorescent staining. In addition, we examined the mRNA expression of angiogenin in HUVEC before and after BDNF stimulation by quantitative reverse transcriptase-polymerase chain reaction. As a result, we noted that BDNF induced angiogenin secretion and nuclear translocation without an increase in the mRNA expression in HUVEC. Furthermore, we demonstrated that BDNF-induced HUVEC proliferation was significantly suppressed when neomycin, a specific inhibitor of nuclear translocation of angiogenin, was administered. These findings indicate that nuclear translocation of angiogenin is critically involved in BDNF-induced proliferation of HUVEC. In conclusion, angiogenin contributes to angiogenesis induced by BDNF.
Collapse
Affiliation(s)
- Ayako Mori
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yusuke Nishioka
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Mai Yamada
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yuka Nishibata
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoyuki Honma
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | | | - Akihiro Ishizu
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
42
|
Totoson P, Pedard M, Marie C, Demougeot C. Activation of endothelial TrkB receptors induces relaxation of resistance arteries. Vascul Pharmacol 2018; 106:46-53. [PMID: 29471140 DOI: 10.1016/j.vph.2018.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 02/07/2018] [Accepted: 02/17/2018] [Indexed: 12/18/2022]
Abstract
While brain-derived neurotrophic factor (BDNF) was previously reported to induce relaxation of conduit artery, whether the BDNF/TrkB (tropomyosin-related kinase) pathway is involved in the tone control of resistance arteries is not known. This study investigated TrkB receptors levels/localization and the vasomotor effect of the TrkB receptor agonist LM22A-4 in isolated third-order mesenteric arteries from rats. Immunostaining revealed the presence of both full-length and truncated TrkB receptors, especially at the endothelial level. By using wire myography, LM22A-4 induced vascular relaxation that was significantly decreased by cyclotraxin B as a non-competitive TrkB antagonist and fully prevented by endothelium removal. Inhibitors of NO, EDHF, PGI2 production and the PI3K/Akt pathways separately reduced LM22A-4 induced-relaxation. By contrast, inhibition of Raf/MEK, PLCγ and CaM/CaMKII pathways did not change the relaxant effect of LM22A-4. Interestingly, BDNF also induced an endothelium and TrkB-dependent relaxation. These results indicate that endothelial TrkB activation results in the relaxation of resistance vessels via PI3K/Akt-induced eNOS phosphorylation and production of EDHF and PGI2. These data are consistent with the contribution of the endothelial BDNF/TrkB pathway to the regulation of peripheral vascular tone. They also validate the use of LM22A-4 as a reliable pharmacological agent for studying the vascular effect of BDNF.
Collapse
Affiliation(s)
- P Totoson
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030 Besançon, France
| | - M Pedard
- INSERM UMR 1093-CAPS, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France
| | - C Marie
- INSERM UMR 1093-CAPS, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - C Demougeot
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
43
|
Pedard M, Quirié A, Garnier P, Tessier A, Demougeot C, Marie C. The Cerebral Brain-Derived Neurotrophic Factor Pathway, Either Neuronal or Endothelial, Is Impaired in Rats with Adjuvant-Induced Arthritis. Connection with Endothelial Dysfunction. Front Physiol 2018; 8:1125. [PMID: 29375397 PMCID: PMC5767301 DOI: 10.3389/fphys.2017.01125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/20/2017] [Indexed: 01/16/2023] Open
Abstract
Cognitive abilities are largely dependent on activation of cerebral tropomyosin-related kinase B receptors (TrkB) by brain-derived neurotrophic factor (BDNF) that is secreted under a bioactive form by both neurons and endothelial cells. In addition, there is mounting evidence for a link between endothelial function and cognition even though the underlying mechanisms are not well known. Therefore, we investigated the cerebral BDNF pathway, either neuronal or endothelial, in rheumatoid arthritis (RA) that combines both endothelial dysfunction (ED) and impaired cognition. Adjuvant-induced arthritis (AIA) in rats was used as a model of RA. Clinical inflammatory symptoms were evaluated from an arthritis score and brains were collected at day 31 ± 2 post-immunization. Neuronal expression of BDNF and TrkB phosphorylated at tyrosine 816 (p-TrkB) was examined in brain slices. Endothelial BDNF and p-TrkB expression was examined on both brain slices (hippocampal arterioles) and isolated cerebral microvessels-enriched fractions (vessels downstream to arterioles). The connection between endothelial nitric oxide (NO) and BDNF production was explored on the cerebrovascular fractions using endothelial NO synthase (eNOS) levels as a marker of NO production, Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME) as a NOS inhibitor and glyceryl-trinitrate as a slow releasing NO donor. Brain slices displayed lower BDNF and p-TrkB staining in both neurons and arteriolar endothelial cells in AIA than in control rats. For endothelial cells but not neurons, a strong correlation was observed between BDNF and p-TrkB staining. Of note, a strong correlation was also observed between neuronal p-TrkB and endothelial BDNF staining. In cerebral microvessels-enriched fractions, AIA led to decreased BDNF and eNOS levels with a positive association between the 2 parameters. These effects coincided with decreased BDNF and p-TrkB staining in endothelial cells. The exposure of AIA cerebrovascular fractions to GTN increased BDNF levels while the exposure of control fractions to L-NAME decreased BDNF levels. Changes in the cerebral BDNF pathway were not associated with arthritis score. The present study reveals that AIA impairs the endothelial and neuronal BDNF/TrkB pathway, irrespective of the severity of inflammatory symptoms but dependent on endothelial NO production. These results open new perspectives for the understanding of the link between ED and impaired cognition.
Collapse
Affiliation(s)
- Martin Pedard
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France.,Service de Neurologie, CHRU Dijon, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| | - Anne Tessier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| | - Céline Demougeot
- EA4267 PEPITE, FHU INCREASE, University of Bourgogne Franche-Comté, Besançon, France
| | - Christine Marie
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| |
Collapse
|
44
|
Lazarovici P, Lahiani A, Gincberg G, Haham D, Fluksman A, Benny O, Marcinkiewicz C, Lelkes PI. Nerve Growth Factor-Induced Angiogenesis: 1. Endothelial Cell Tube Formation Assay. Methods Mol Biol 2018; 1727:239-250. [PMID: 29222786 DOI: 10.1007/978-1-4939-7571-6_18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Nerve growth factor (NGF) is a neurotrophin promoting survival, proliferation, differentiation, and neuroprotection in the embryonal and adult nervous system. NGF also induces angiogenic effects in the cardiovascular system, which may be beneficial in engineering new blood vessels and for developing novel anti-angiogenesis therapies for cancer. Angiogenesis is a cellular process characterized by a number of events, including endothelial cell migration, invasion, and assembly into capillaries. In vitro endothelial tube formation assays are performed using primary human umbilical vein endothelial cells, human aortic endothelial cells, and other human or rodent primary endothelial cells isolated from the vasculature of both tumors and normal tissues. Immortalized endothelial cell lines are also used for these assays. When seeded onto Matrigel, these cells reorganize to create tubelike structure, which may be used as models for studying some aspects of in vitro angiogenesis. Image acquisition by light and fluorescence microscopy and/or quantification of fluorescently labeled cells can be carried out manually or digitally, using commercial software and automated image processing. Here we detail materials, procedure, assay conditions, and cell labeling for quantification of endothelial cell tube formation. This model can be applied to study cellular and molecular mechanisms by which NGF or other neurotrophins promote angiogenesis. This model may also be useful for the development of potential angiogenic and/or anti-angiogenic drugs targeting NGF receptors.
Collapse
Affiliation(s)
- Philip Lazarovici
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Adi Lahiani
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Galit Gincberg
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dikla Haham
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arnon Fluksman
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Benny
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Cezary Marcinkiewicz
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, USA
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, USA
| |
Collapse
|
45
|
Hu L, Qi S, Zhang K, Fu Q. Essential role of brain-derived neurotrophic factor (bdnf) in diabetic erectile dysfunction. Andrologia 2017; 50. [PMID: 29159909 DOI: 10.1111/and.12924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2017] [Indexed: 01/09/2023] Open
Affiliation(s)
- L. Hu
- Department of Urology; Shandong Provincial Hospital Affiliated to Shandong University; Jinan China
- Department of Urology; Shandong Zaozhuang Municipal Hospital; Zaozhuang China
| | - S. Qi
- Department of Neurology; Shandong Zaozhuang Municipal Hospital; Zaozhuang China
| | - K. Zhang
- Department of Urology; Shandong Provincial Hospital Affiliated to Shandong University; Jinan China
| | - Q. Fu
- Department of Urology; Shandong Provincial Hospital Affiliated to Shandong University; Jinan China
| |
Collapse
|
46
|
Li B, Gao Y, Zhang W, Xu JR. Regulation and effects of neurotrophic factors after neural stem cell transplantation in a transgenic mouse model of Alzheimer disease. J Neurosci Res 2017; 96:828-840. [PMID: 29114922 DOI: 10.1002/jnr.24187] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 12/14/2022]
Abstract
According to much research, neurodegeneration and cognitive decline in Alzheimer disease (AD) are correlated with alternations of neurotrophic factors such as nerve growth factor, brain-derived neurotrophic factor, and glial cell-derived neurotrophic factor. The experimental illumination of neural stem cell (NSC) transplantation to eliminate AD symptoms is being explored frequently, and we have acknowledged that neurotrophic factors may play a pivotal role in cognitive improvement. However, the relation between the reversal of cognitive deficits after NSC transplantation and directed alternations of neurotrophic factors is not clearly expounded. Meanwhile, reduced inflammatory response, promoted vessel density, and vascular endothelial growth factor (VEGF) can be reflections of improvement in cerebrovascular function. Three weeks after NSC transplantation, spatial learning and memory function in NSC-injected (Tg-NSC) mice were significantly improved compared with vehicle-injected (Tg-Veh) mice. Meanwhile, results obtained by immunofluorescence and Western blot analyses demonstrated that the levels of neurotrophic factors, VEGF, and vessel density in the cortex of Tg-NSC mice were significantly enhanced compared with Tg-Veh mice, while the levels of proinflammatory cytokines interleukin (IL)-1β, tumor necrosis factor-α, and IL-6 were significantly decreased. Our results suggest that elevated concentrations of neurotrophic factors probably play a critical role in rescuing cognitive dysfunction in APP/PS1 transgenic mice after NSC transplantation, and neurotrophic factors may improve cerebrovascular function by means such as reducing inflammatory response and promoting angiogenesis.
Collapse
Affiliation(s)
- Bo Li
- Department of Medical Imaging, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yun Gao
- Department of Medical Imaging, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Wei Zhang
- Department of Medical Imaging, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jian-Rong Xu
- Department of Medical Imaging, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|
47
|
Pak RW, Hadjiabadi DH, Senarathna J, Agarwal S, Thakor NV, Pillai JJ, Pathak AP. Implications of neurovascular uncoupling in functional magnetic resonance imaging (fMRI) of brain tumors. J Cereb Blood Flow Metab 2017; 37:3475-3487. [PMID: 28492341 PMCID: PMC5669348 DOI: 10.1177/0271678x17707398] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Functional magnetic resonance imaging (fMRI) serves as a critical tool for presurgical mapping of eloquent cortex and changes in neurological function in patients diagnosed with brain tumors. However, the blood-oxygen-level-dependent (BOLD) contrast mechanism underlying fMRI assumes that neurovascular coupling remains intact during brain tumor progression, and that measured changes in cerebral blood flow (CBF) are correlated with neuronal function. Recent preclinical and clinical studies have demonstrated that even low-grade brain tumors can exhibit neurovascular uncoupling (NVU), which can confound interpretation of fMRI data. Therefore, to avoid neurosurgical complications, it is crucial to understand the biophysical basis of NVU and its impact on fMRI. Here we review the physiology of the neurovascular unit, how it is remodeled, and functionally altered by brain cancer cells. We first discuss the latest findings about the components of the neurovascular unit. Next, we synthesize results from preclinical and clinical studies to illustrate how brain tumor induced NVU affects fMRI data interpretation. We examine advances in functional imaging methods that permit the clinical evaluation of brain tumors with NVU. Finally, we discuss how the suppression of anomalous tumor blood vessel formation with antiangiogenic therapies can "normalize" the brain tumor vasculature, and potentially restore neurovascular coupling.
Collapse
Affiliation(s)
- Rebecca W Pak
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Darian H Hadjiabadi
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Janaka Senarathna
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Shruti Agarwal
- 2 Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Nitish V Thakor
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jay J Pillai
- 2 Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Arvind P Pathak
- 1 Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA.,2 Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, USA.,3 Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
48
|
Frank AP, Palmer BF, Clegg DJ. Do estrogens enhance activation of brown and beiging of adipose tissues? Physiol Behav 2017; 187:24-31. [PMID: 28988965 DOI: 10.1016/j.physbeh.2017.09.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/30/2017] [Accepted: 09/30/2017] [Indexed: 01/05/2023]
Abstract
Obesity and its associated co-morbidities are worldwide public health concerns. Obesity is characterized by excessive adipose tissue accumulation; however, it is important to recognize that human and rodent adipose tissues are made up of several distinct adipose tissue sub-types. White adipose tissue (WAT) is considered the prototypical fat cell, due to its capacity and capability to store large amounts of lipid. In contrast, brown adipose tissue (BAT) oxidizes substrates to generate heat. BAT contains more mitochondria than WAT and express uncoupling protein-1 (UCP1), which mediates BAT thermogenesis. A third sub-type of adipose tissue, Brown-in-white (BRITE)/beige adipocytes arise from WAT upon adrenergic stimulation and resembles BAT functionally. The energy burning feature of BAT/beige cells, combined with evidence of an inverse-correlation between BAT/beige adipose tissue and obesity have given rise to the hypothesis that obesity may be linked to BAT/beige 'malfunction'. Females have more BAT and perhaps an enhanced capacity to beige their adipose tissue when compared to males. Multiple signal pathways are capable of activating BAT thermogenesis and beiging of WAT; here, we discuss the potential role of estrogens in enhancing and mediating these factors to enhance adipose tissue thermogenesis.
Collapse
Affiliation(s)
- Aaron P Frank
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Deborah J Clegg
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
49
|
Wu KW, Mo JL, Kou ZW, Liu Q, Lv LL, Lei Y, Sun FY. Neurovascular Interaction Promotes the Morphological and Functional Maturation of Cortical Neurons. Front Cell Neurosci 2017; 11:290. [PMID: 28966577 PMCID: PMC5605567 DOI: 10.3389/fncel.2017.00290] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/04/2017] [Indexed: 01/19/2023] Open
Abstract
Brain microvascular endothelial cells (BMEC) have been found to guide the migration, promote the survival and regulate the differentiation of neural cells. However, whether BMEC promote development and maturation of immature neurons is still unknown. Therefore, in this study, we used a direct endothelium-neuron co-culture system combined with patch clamp recordings and confocal imaging analysis, to investigate the effects of endothelial cells on neuronal morphology and function during development. We found that endothelial cells co-culture or BMEC-conditioned medium (B-CM) promoted neurite outgrowth and spine formation, accelerated electrophysiological development and enhanced synapse function. Moreover, B-CM treatment induced vascular endothelial growth factor (VEGF) expression and p38 phosphorylation in the cortical neurons. Through pharmacological analysis, we found that incubation with SU1498, an inhibitor of VEGF receptor, abolished B-CM-induced p-p38 upregulation and suppressed the enhancement of synapse formation and transmission. SB203580, an inhibitor of p38 MAPK also blocked B-CM-mediated synaptic regulation. Together these results clearly reveal that the endothelium-neuron interactions promote morphological and functional maturation of neurons. In addition, neurovascular interaction-mediated promotion of neural network maturation relies on activation of VEGF/Flk-1/p38 MAPK signaling. This study provides novel aspects of endothelium-neuron interactions and novel mechanism of neurovascular crosstalk.
Collapse
Affiliation(s)
- Kun-Wei Wu
- Institute of Biomedical Sciences and Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China
| | - Jia-Lin Mo
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Zeng-Wei Kou
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Qi Liu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Ling-Ling Lv
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Yu Lei
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Feng-Yan Sun
- Institute of Biomedical Sciences and Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| |
Collapse
|
50
|
Pius-Sadowska E, Machaliński B. BDNF - A key player in cardiovascular system. J Mol Cell Cardiol 2017; 110:54-60. [PMID: 28736262 DOI: 10.1016/j.yjmcc.2017.07.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
Neurotrophins (NTs) were first identified as target-derived survival factors for neurons of the central and peripheral nervous system (PNS). They are known to control neural cell fate, development and function. Independently of their neuronal properties, NTs exert unique cardiovascular activity. The heart is innervated by sensory, sympathetic and parasympathetic neurons, which require NTs during early development and in the establishment of mature properties, contributing to the maintenance of cardiovascular homeostasis. The identification of molecular mechanisms regulated by NTs and involved in the crosstalk between cardiac sympathetic nerves, cardiomyocytes, cardiac fibroblasts, and vascular cells, has a fundamental importance in both normal heart function and disease. The article aims to review the recent data on the effects of Brain-Derived Neurotrophic Factor (BDNF) on various cardiovascular neuronal and non-neuronal functions such as the modulation of synaptic properties of autonomic neurons, axonal outgrowth and sprouting, formation of the vascular and neural networks, smooth muscle migration, and control of endothelial cell survival and cardiomyocytes. Understanding these mechanisms may be crucial for developing novel therapeutic strategies, including stem cell-based therapies.
Collapse
Affiliation(s)
- Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|