1
|
Cai B, Fang J, Zhou S, Xie M, Zhang K, Li J, Yin G. Enzyme-crosslinked hyaluronic acid hydrogel scaffolds for BMSCs microenvironment and wound healing. Int J Biol Macromol 2025; 295:139566. [PMID: 39788243 DOI: 10.1016/j.ijbiomac.2025.139566] [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: 09/16/2024] [Revised: 12/17/2024] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
Tissue engineering utilizing hydrogel scaffolds in combination with exogenous stem cells holds significant potential for promoting wound regeneration. However, the microenvironment provided by existing skin tissue engineering scaffold materials is often inadequate. Herein, we demonstrate an enzyme-crosslinked hyaluronic acid hydrogel to provide a growth microenvironment for exogenous bone marrow mesenchymal stem cells and promote acute wound healing. This material is developed by grafting dopamine onto hyaluronic acid, followed by enzyme crosslinking using horseradish peroxidase and hydrogen peroxide, which creates a loose, porous structure. The hydrogel possesses adhesive and self-healing properties, offering a microenvironment with excellent cell compatibility for exogenous BMSCs. In vivo studies showed that this hydrogel significantly accelerated the healing of acute full-thickness skin wounds, resulting in the formation of appendages such as hair follicles and minimal scarring. This study not only presents a novel skin tissue engineering scaffold but also offers a promising clinical strategy for achieving scar-minimized wound healing.
Collapse
Affiliation(s)
- Bingjie Cai
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jiaao Fang
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shengxi Zhou
- School of Life Science, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mengbo Xie
- School of Life Science, Zhengzhou University, Zhengzhou 450001, PR China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Guangwen Yin
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
2
|
Zhu S, Wu Q, Ying Y, Mao Y, Lu W, Xu J, Cai X, He H, Wu J. Tissue-Adaptive BSA Hydrogel with Dual Release of PTX and bFGF Promotes Spinal Cord Injury Repair via Glial Scar Inhibition and Axon Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401407. [PMID: 39385643 DOI: 10.1002/smll.202401407] [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: 02/22/2024] [Revised: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Spinal cord injury (SCI) is a severe clinical disease usually accompanied by activated glial scar, neuronal axon rupture, and disabled motor function. To mimic the microenvironment of the SCI injury site, a hydrogel system with a comparable mechanical property to the spinal cord is desirable. Therefore, a novel elastic bovine serum albumin (BSA) hydrogel is fabricated with excellent adhesive, injectable, and biocompatible properties. The hydrogel is used to deliver paclitaxel (PTX) together with basic fibroblast growth factor (bFGF) to inhibit glial scar formation as well as promote axon regeneration and motor function for SCI repair. Due to the specific interaction of BSA with both drugs, bFGF, and PTX can be controllably released from the hydrogel system to achieve an effective concentration at the wound site during the SCI regeneration process. Moreover, benefiting from the combination of PTX and bFGF, this bFGF/PTX@BSA system significantly aided axon repair by promoting the elongation of axons across the glial scar with reduced reactive astrocyte secretion. In addition, remarkable anti-apoptosis of nerve cells is evident with the bFGF/PTX@BSA system. Subsequently, this multi-functionalized drug system significantly improved the motor function of the rats after SCI. These results reveal that bFGF/PTX@BSA is an ideal functionalized material for nerve repair in SCI.
Collapse
Affiliation(s)
- Sipin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Qiuji Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yibo Ying
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuqin Mao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenjie Lu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiong Cai
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huacheng He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Jiang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
3
|
Scopa C, Barnada SM, Cicardi ME, Singer M, Trotti D, Trizzino M. JUN upregulation drives aberrant transposable element mobilization, associated innate immune response, and impaired neurogenesis in Alzheimer's disease. Nat Commun 2023; 14:8021. [PMID: 38049398 PMCID: PMC10696058 DOI: 10.1038/s41467-023-43728-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 11/06/2023] [Indexed: 12/06/2023] Open
Abstract
Adult neurogenic decline, inflammation, and neurodegeneration are phenotypic hallmarks of Alzheimer's disease (AD). Mobilization of transposable elements (TEs) in heterochromatic regions was recently reported in AD, but the underlying mechanisms are still underappreciated. Combining functional genomics with the differentiation of familial and sporadic AD patient derived-iPSCs into hippocampal progenitors, CA3 neurons, and cerebral organoids, we found that the upregulation of the AP-1 subunit, c-Jun, triggers decondensation of genomic regions containing TEs. This leads to the cytoplasmic accumulation of HERVK-derived RNA-DNA hybrids, the activation of the cGAS-STING cascade, and increased levels of cleaved caspase-3, suggesting the initiation of programmed cell death in AD progenitors and neurons. Notably, inhibiting c-Jun effectively blocks all these downstream molecular processes and rescues neuronal death and the impaired neurogenesis phenotype in AD progenitors. Our findings open new avenues for identifying therapeutic strategies and biomarkers to counteract disease progression and diagnose AD in the early, pre-symptomatic stages.
Collapse
Affiliation(s)
- Chiara Scopa
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Samantha M Barnada
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria E Cicardi
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mo Singer
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Marco Trizzino
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
4
|
Zhang S, Zhang Y, Zheng Y, Zhu S, Sun J, Deng Y, Wang Q, Zhai Q. Dexmedetomidine attenuates sleep deprivation-induced inhibition of hippocampal neurogenesis via VEGF-VEGFR2 signaling and inhibits neuroinflammation. Biomed Pharmacother 2023; 165:115085. [PMID: 37392656 DOI: 10.1016/j.biopha.2023.115085] [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: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023] Open
Abstract
Long periods of sleep deprivation (SD) have serious effects on health. While the α2 adrenoceptor agonist dexmedetomidine (DEX) can improve sleep quality for patients who have insomnia, the effect of DEX on cognition and mechanisms after SD remains elusive. C57BL/6 mice were subjected to 20 h SD daily for seven days. DEX (100 μg/kg) was administered intravenously twice daily (at 1:00 p.m. and 3:00 p.m.) during seven days of SD. We found that systemic administration of DEX attenuated cognitive deficits by performing the Y maze and novel object recognition tests and increased DCX+, SOX2+, Ki67+, and BrdU+NeuN+/NeuN+ cell numbers in the dentate gyrus (DG) region of SD mice by using immunofluorescence, western blotting, and BrdU staining. DEX did not reverse the decrease in DCX+, SOX2+, or Ki67+ cell numbers in SD mice after administration of the α2A-adrenoceptor antagonist BRL-44408. Furthermore, the vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) expression was upregulated in SD+DEX mice compared with SD mice. Luminex analysis showed that the neurogenic effects of DEX were possibly related to the inhibition of neuroinflammation, including IL-1α, IL-2, CCL5, and CXCL1. Our results suggested that DEX alleviated the impaired learning and memory of SD mice potentially by inducing hippocampal neurogenesis via the VEGF-VEGFR2 signaling pathway and by suppressing neuroinflammation, and α2A adrenoceptors are required for the neurogenic effects of DEX after SD. This novel mechanism may add to our knowledge of DEX in the clinical treatment of impaired memory caused by SD.
Collapse
Affiliation(s)
- Shuyue Zhang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ying Zhang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Yige Zheng
- The Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, Shaanxi, China
| | - Shan Zhu
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Jianyu Sun
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Yingying Deng
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Qian Zhai
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
5
|
Radoszkiewicz K, Hribljan V, Isakovic J, Mitrecic D, Sarnowska A. Critical points for optimizing long-term culture and neural differentiation capacity of rodent and human neural stem cells to facilitate translation into clinical settings. Exp Neurol 2023; 363:114353. [PMID: 36841464 DOI: 10.1016/j.expneurol.2023.114353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/03/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023]
Abstract
Despite several decades of research on the nature and functional properties of neural stem cells, which brought great advances in regenerative medicine, there is still a plethora of ambiguous protocols and interpretations linked to their applications. Here, we present a whole spectrum of protocol elements that should be standardized in order to obtain viable cell cultures and facilitate their translation into clinical settings. Additionally, this review also presents outstanding limitations and possible problems to be encountered when dealing with protocol optimization. Most importantly, we also outline the critical points that should be considered before starting any experiments utilizing neural stem cells or interpreting their results.
Collapse
Affiliation(s)
- Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, 02-106 Warsaw, Poland
| | - Valentina Hribljan
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, Croatia
| | - Jasmina Isakovic
- Omnion Research International Ltd, Heinzelova 4, 10000 Zagreb, Croatia
| | - Dinko Mitrecic
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, Croatia
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, 02-106 Warsaw, Poland.
| |
Collapse
|
6
|
Hoseinzadeh A, Ghoddusi Johari H, Anbardar MH, Tayebi L, Vafa E, Abbasi M, Vaez A, Golchin A, Amani AM, Jangjou A. Effective treatment of intractable diseases using nanoparticles to interfere with vascular supply and angiogenic process. Eur J Med Res 2022; 27:232. [PMID: 36333816 PMCID: PMC9636835 DOI: 10.1186/s40001-022-00833-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is a vital biological process involving blood vessels forming from pre-existing vascular systems. This process contributes to various physiological activities, including embryonic development, hair growth, ovulation, menstruation, and the repair and regeneration of damaged tissue. On the other hand, it is essential in treating a wide range of pathological diseases, such as cardiovascular and ischemic diseases, rheumatoid arthritis, malignancies, ophthalmic and retinal diseases, and other chronic conditions. These diseases and disorders are frequently treated by regulating angiogenesis by utilizing a variety of pro-angiogenic or anti-angiogenic agents or molecules by stimulating or suppressing this complicated process, respectively. Nevertheless, many traditional angiogenic therapy techniques suffer from a lack of ability to achieve the intended therapeutic impact because of various constraints. These disadvantages include limited bioavailability, drug resistance, fast elimination, increased price, nonspecificity, and adverse effects. As a result, it is an excellent time for developing various pro- and anti-angiogenic substances that might circumvent the abovementioned restrictions, followed by their efficient use in treating disorders associated with angiogenesis. In recent years, significant progress has been made in different fields of medicine and biology, including therapeutic angiogenesis. Around the world, a multitude of research groups investigated several inorganic or organic nanoparticles (NPs) that had the potential to effectively modify the angiogenesis processes by either enhancing or suppressing the process. Many studies into the processes behind NP-mediated angiogenesis are well described. In this article, we also cover the application of NPs to encourage tissue vascularization as well as their angiogenic and anti-angiogenic effects in the treatment of several disorders, including bone regeneration, peripheral vascular disease, diabetic retinopathy, ischemic stroke, rheumatoid arthritis, post-ischemic cardiovascular injury, age-related macular degeneration, diabetic retinopathy, gene delivery-based angiogenic therapy, protein delivery-based angiogenic therapy, stem cell angiogenic therapy, and diabetic retinopathy, cancer that may benefit from the behavior of the nanostructures in the vascular system throughout the body. In addition, the accompanying difficulties and potential future applications of NPs in treating angiogenesis-related diseases and antiangiogenic therapies are discussed.
Collapse
Affiliation(s)
- Ahmad Hoseinzadeh
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Ghoddusi Johari
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Ehsan Vafa
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Golchin
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Choi JK, Kwak IS, Yoon SB, Cho H, Moon BS. A Small Molecule Promoting Neural Differentiation Suppresses Cancer Stem Cells in Colorectal Cancer. Biomedicines 2022; 10:biomedicines10040859. [PMID: 35453609 PMCID: PMC9025482 DOI: 10.3390/biomedicines10040859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer stem cells (CSCs) are a tumor cell subpopulation that drives tumor progression and metastasis, leading to a poor overall survival of patients. In colorectal cancer (CRC), the hyper-activation of Wnt/β-catenin signaling by a mutation of both adenomatous polyposis coli (APC) and K-Ras increases the size of the CSC population. We previously showed that CPD0857 inactivates Wnt/β-catenin signaling by promoting the ubiquitin-dependent proteasomal degradation of β-catenin and Ras proteins, thereby decreasing proliferation and increasing the apoptosis of CRC lines. CPD0857 also decreased the growth and invasiveness of CRC cells harboring mutant K-Ras resistant to EGFR mAb therapy. Here, we show that CPD0857 treatment decreases proliferation and increases the neuronal differentiation of neural progenitor cells (NPCs). CDP0857 effectively reduced the expression of CSC markers and suppressed self-renewal capacity. CPD0857 treatment also inhibited the proliferation and expression of CSC markers in D-K-Ras MT cells carrying K-Ras, APC and PI3K mutations, indicating the inhibition of PI3K/AKT signaling. Moreover, CPD0857-treated xenograft mice showed a regression of tumor growth and decreased numbers of CSCs in tumors. We conclude that CPD0857 could serve as the basis of a drug development strategy targeting CSCs activated through Wnt/β-catenin-Ras MAPK-PI3K/AKT signaling in CRCs.
Collapse
Affiliation(s)
- Jung Kyu Choi
- Department of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Korea;
| | - Ihn-Sil Kwak
- Department of Ocean Integrated Science, Chonnam National University, Yeosu 59626, Korea;
| | - Sae-Bom Yoon
- Therapeutics and Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (S.-B.Y.); (H.C.)
| | - Heeyeong Cho
- Therapeutics and Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (S.-B.Y.); (H.C.)
| | - Byoung-San Moon
- Department of Biotechnology, Chonnam National University, Yeosu 59626, Korea
- Correspondence: ; Tel.: +82-61-659-7307; Fax: +82-61-659-7309
| |
Collapse
|
8
|
Jing J, Jiang H, Zhang L. Endothelial progenitor cells promote neural stem cell proliferation in hypoxic conditions through VEGF via the PI3K/AKT pathway. J Recept Signal Transduct Res 2022; 42:479-485. [PMID: 35042445 DOI: 10.1080/10799893.2021.2019275] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neurons and vascular cells compose neurovascular niches in the central nervous system where endothelial cells can promote neurogenesis via direct and indirect effects. Neurocytes and vascular cells are gravely destroyed upon spinal cord injury, which severely affects spinal motor functions. Neurogenesis originates from neural stem cells (NSCs) and endothelial cells derived from endothelial progenitor cells (EPCs) in the spinal cord. To demonstrate whether EPCs promote NSC proliferation, we cultured NSCs with EPC-conditioned medium from hypoxic conditions (CM) and EPC-unconditioned medium (UCM), i.e. endothelial cell basal medium-2, as a control. The number of S-phase cells in CM were 54.73 ± 0.67 whereas those in UCM were 26.30 ± 0.43, and the number of cells in CM was higher than that in UCM (0.32 ± 0.0019 vs. 0.55 ± 0.0029). We hypothesized that the cell proliferation was promoted by vascular endothelial growth factor A (VEGFA), which is secreted by EPCs in hypoxic conditions. We then used VEGF shRNA to decrease VEGFA secretion by EPCs. NSCs were cultured in conditioned medium from shRNA transfected EPCs under hypoxia (shRNA-CM) and EPC-conditioned medium under hypoxia (CM). The number of S-phase cells in the shRNA-CM was 36.86 ± 0.49 whereas that in CM was 53.61 ± 0.89, and the number of cells in the shRNA-CM was lower than that in CM (0.55 ± 0.0032 vs. 0.34 ± 0.0029). These data indicate that EPCs could promote NSC proliferation in hypoxic condition through VEGFA secretion.
Collapse
Affiliation(s)
- Jingti Jing
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haoming Jiang
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lin Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
9
|
The interplay of neurovasculature and adult hippocampal neurogenesis. Neurosci Lett 2021; 760:136071. [PMID: 34147540 DOI: 10.1016/j.neulet.2021.136071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/06/2021] [Accepted: 06/15/2021] [Indexed: 01/14/2023]
Abstract
The subgranular zone of the dentate gyrus provides a local microenvironment (niche) for neural stem cells. In the adult brain, it has been established that the vascular compartment of such niches has a significant role in regulating adult hippocampal neurogenesis. More recently, evidence showed that neurovascular coupling, the relationship between blood flow and neuronal activity, also regulates hippocampal neurogenesis. Here, we review the most recent articles on addressing the intricate relationship between neurovasculature and adult hippocampal neurogenesis and a novel pathway where functional hyperemia enhances hippocampal neurogenesis. In the end, we have further reviewed recent research showing that impaired neurovascular coupling may cause declined neurogenesis and contribute to brain damage in neurodegenerative diseases.
Collapse
|
10
|
Therapeutic Nanoparticles for the Different Phases of Ischemic Stroke. Life (Basel) 2021; 11:life11060482. [PMID: 34073229 PMCID: PMC8227304 DOI: 10.3390/life11060482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/27/2022] Open
Abstract
Stroke represents the second leading cause of mortality and morbidity worldwide. Ischemic strokes are the most prevalent type of stroke, and they are characterized by a series of pathological events prompted by an arterial occlusion that leads to a heterogeneous pathophysiological response through different hemodynamic phases, namely the hyperacute, acute, subacute, and chronic phases. Stroke treatment is highly reliant on recanalization therapies, which are limited to only a subset of patients due to their narrow therapeutic window; hence, there is a huge need for new stroke treatments. Nonetheless, the vast majority of promising treatments are not effective in the clinical setting due to their inability to cross the blood-brain barrier and reach the brain. In this context, nanotechnology-based approaches such as nanoparticle drug delivery emerge as the most promising option. In this review, we will discuss the current status of nanotechnology in the setting of stroke, focusing on the diverse available nanoparticle approaches targeted to the different pathological and physiological repair mechanisms involved in each of the stroke phases.
Collapse
|
11
|
Winkelman MA, Koppes AN, Koppes RA, Dai G. Bioengineering the neurovascular niche to study the interaction of neural stem cells and endothelial cells. APL Bioeng 2021; 5:011507. [PMID: 33688617 PMCID: PMC7932757 DOI: 10.1063/5.0027211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
The ability of mammalian neural stem cells (NSCs) to self-renew and differentiate throughout adulthood has made them ideal to study neurogenesis and attractive candidates for neurodegenerative disease therapies. In the adult mammalian brain, NSCs are maintained in the neurovascular niche (NVN) where they are found near the specialized blood vessels, suggesting that brain endothelial cells (BECs) are prominent orchestrators of NSC fate. However, most of the current knowledge of the mammalian NVN has been deduced from nonhuman studies. To circumvent the challenges of in vivo studies, in vitro models have been developed to better understand the reciprocal cellular mechanisms of human NSCs and BECs. This review will cover the current understanding of mammalian NVN biology, the effects of endothelial cell-derived signals on NSC fate, and the in vitro models developed to study the interactions between NSCs and BECs.
Collapse
Affiliation(s)
- Max A Winkelman
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| | | | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| |
Collapse
|
12
|
Chan SJ, Esposito E, Hayakawa K, Mandaville E, Smith RAA, Guo S, Niu W, Wong PTH, Cool SM, Lo EH, Nurcombe V. Vascular Endothelial Growth Factor 165-Binding Heparan Sulfate Promotes Functional Recovery From Cerebral Ischemia. Stroke 2020; 51:2844-2853. [PMID: 32772683 DOI: 10.1161/strokeaha.119.025304] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND PURPOSE Although VEGF165 (vascular endothelial growth factor-165) is able to enhance both angiogenesis and neurogenesis, it also increases vascular permeability through the blood-brain barrier. Heparan sulfate (HS) sugars play important roles in regulating VEGF bioactivity in the pericellular compartment. Here we asked whether an affinity-purified VEGF165-binding HS (HS7) could augment endogenous VEGF activity during stroke recovery without affecting blood-brain barrier function. METHODS Both rat brain endothelial cell line 4 and primary rat neural progenitor cells were used to evaluate the potential angiogenic and neurogenic effects of HS7 in vitro. For in vivo experiments, male Sprague-Dawley rats were subjected to 100 minutes of transient focal cerebral ischemia, then treated after 4 days with either PBS or HS7. One week later, infarct volume, behavioral sequelae, immunohistochemical markers of angiogenesis and neural stem cell proliferation were assessed. RESULTS HS7 significantly enhanced VEGF165-mediated angiogenesis in rat brain endothelial cell line 4 brain endothelial cells, and increased the proliferation and differentiation of primary neural progenitor cells, both via the VEGFR2 (vascular endothelial growth factor receptor 2) pathway. Intracerebroventricular injection of HS7 improved neurological outcome in ischemic rats without changing infarct volumes. Immunostaining of the compromised cerebrum demonstrated increases in collagen IV/Ki67 and nestin/Ki67 after HS7 exposure, consistent with its ability to promote angiogenesis and neurogenesis, without compromising blood-brain barrier integrity. CONCLUSIONS A VEGF-activating glycosaminoglycan sugar, by itself, is able to enhance endogenous VEGF165 activity during the post-ischemic recovery phase of stroke.
Collapse
Affiliation(s)
- Su Jing Chan
- Department of Radiology (S.J.C., E.E., K.H., E.M., S.G., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown.,Institute of Medical Biology, Glycotherapeutics Group, A*STAR (S.J.C., R.A.A.S., S.M.C., V.N.)
| | - Elga Esposito
- Department of Radiology (S.J.C., E.E., K.H., E.M., S.G., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Kazuhide Hayakawa
- Department of Radiology (S.J.C., E.E., K.H., E.M., S.G., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown.,Department of Neurology (K.H., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Emiri Mandaville
- Department of Radiology (S.J.C., E.E., K.H., E.M., S.G., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Raymond A A Smith
- Institute of Medical Biology, Glycotherapeutics Group, A*STAR (S.J.C., R.A.A.S., S.M.C., V.N.)
| | - Shuzhen Guo
- Department of Radiology (S.J.C., E.E., K.H., E.M., S.G., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Wanting Niu
- Tissue Engineering Laboratories, VA Boston Healthcare System, MA (W.N.)
| | | | - Simon M Cool
- Institute of Medical Biology, Glycotherapeutics Group, A*STAR (S.J.C., R.A.A.S., S.M.C., V.N.)
| | - Eng H Lo
- Department of Radiology (S.J.C., E.E., K.H., E.M., S.G., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown.,Department of Neurology (K.H., E.H.L.), Massachusetts General Hospital, Harvard Medical School, Charlestown
| | - Victor Nurcombe
- Institute of Medical Biology, Glycotherapeutics Group, A*STAR (S.J.C., R.A.A.S., S.M.C., V.N.)
| |
Collapse
|
13
|
Adult Neurogenesis in the Subventricular Zone and Its Regulation After Ischemic Stroke: Implications for Therapeutic Approaches. Transl Stroke Res 2019; 11:60-79. [DOI: 10.1007/s12975-019-00717-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
|
14
|
Qu M, Pan J, Wang L, Zhou P, Song Y, Wang S, Jiang L, Geng J, Zhang Z, Wang Y, Tang Y, Yang GY. MicroRNA-126 Regulates Angiogenesis and Neurogenesis in a Mouse Model of Focal Cerebral Ischemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:15-25. [PMID: 30825669 PMCID: PMC6393705 DOI: 10.1016/j.omtn.2019.02.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 01/09/2023]
Abstract
Studies demonstrate that microRNA-126 plays a critical role in promoting angiogenesis. However, its effects on angiogenesis following ischemic stroke are unclear. Here, we explored the effect of microRNA-126-3p and microRNA-126-5p on angiogenesis and neurogenesis after brain ischemia. We demonstrated that both microRNA (miRNA)-126-3p and microRNA-126-5p increased the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) compared with the scrambled miRNA control (p < 0.05). Transferring microRNA-126 into a mouse middle cerebral artery occlusion model via lentivirus, we found that microRNA-126 overexpression increased the number of CD31+/BrdU+ (5-bromo-2'-deoxyuridine-positive) proliferating endothelial cells and DCX+/BrdU+ neuroblasts in the ischemic mouse brain, improved neurobehavioral outcomes (p < 0.05), and reduced brain atrophy volume (p < 0.05) compared with control mice. Western blot results showed that AKT and ERK signaling pathways were activated in the lentiviral-microRNA-126-treated group (p < 0.05). Both PCR and western blot results demonstrated that tyrosine-protein phosphatase non-receptor type 9 (PTPN9) was decreased in the lentiviral-microRNA-126-treated group (p < 0.05). Dual-luciferase gene reporter assay also showed that PTPN9 was the direct target of microRNA-126-3p and microRNA-126-5p in the ischemic brain. We demonstrated that microRNA-126-3p and microRNA-126-5p promoted angiogenesis and neurogenesis in ischemic mouse brain, and further improved neurobehavioral outcomes. Our mechanistic study further showed that microRNA-126 mediated angiogenesis through directly inhibiting its target PTPN9 and activating AKT and ERK signaling pathways.
Collapse
Affiliation(s)
- Meijie Qu
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jiaji Pan
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liping Wang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Panting Zhou
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaying Song
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Shuhong Wang
- Department of Geriatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Lu Jiang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jieli Geng
- Department of Neurology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhijun Zhang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongting Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaohui Tang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
15
|
Ma Y, Wang K, Pan J, Fan Z, Tian C, Deng X, Ma K, Xia X, Huang Y, Zheng JC. Induced neural progenitor cells abundantly secrete extracellular vesicles and promote the proliferation of neural progenitors via extracellular signal-regulated kinase pathways. Neurobiol Dis 2018; 124:322-334. [PMID: 30528256 DOI: 10.1016/j.nbd.2018.12.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/16/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Neural stem/progenitor cells (NPCs) are known to have potent therapeutic effects in neurological disorders through the secretion of extracellular vesicles (EVs). Despite the therapeutic potentials, the numbers of NPCs are limited in the brain, curbing the further use of EVs in the disease treatment. To overcome the limitation of NPC numbers, we used a three transcription factor (Brn2, Sox2, and Foxg1) somatic reprogramming approach to generate induced NPCs (iNPCs) from mouse fibroblasts and astrocytes. The resulting iNPCs released significantly higher numbers of EVs compared with wild-type NPCs (WT-NPCs). Furthermore, iNPCs-derived EVs (iNPC-EVs) promoted NPC function by increasing the proliferative potentials of WT-NPCs. Characterizations of EV contents through proteomics analysis revealed that iNPC-EVs contained higher levels of growth factor-associated proteins that were predicted to activate the down-stream extracellular signal-regulated kinase (ERK) pathways. As expected, the proliferative effects of iNPC-derived EVs on WT-NPCs can be blocked by an ERK pathway inhibitor. Our data suggest potent therapeutic effects of iNPC-derived EVs through the promotion of NPC proliferation, release of growth factors, and activation of ERK pathways. These studies will help develop highly efficient cell-free therapeutic strategies for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yizhao Ma
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Kaizhe Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Jiabin Pan
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Zhaohuan Fan
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Changhai Tian
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China; Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA
| | - Xiaobei Deng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Kangmu Ma
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China; Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China.
| | - Yunlong Huang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China; Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA.
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China; Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China; Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA.
| |
Collapse
|
16
|
Spinal cord organogenesis model reveals role of Flk1 + cells in self-organization of neural progenitor cells into complex spinal cord tissue. Stem Cell Res 2018; 33:156-165. [PMID: 30368192 DOI: 10.1016/j.scr.2018.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/02/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022] Open
Abstract
A platform for studying spinal cord organogenesis in vivo where embryonic stem cell (ESC)-derived neural progenitor cells (NPC) self-organize into spinal cord-like tissue after transplantation in subarachnoid space of the spinal cord has been described. We advance the applicability of this platform by imaging in vivo the formed graft through T2w magnetic resonance imaging (MRI). Furthermore, we used diffusion tensor imaging (DTI) to verify the stereotypical organization of the graft showing that it mimics the host spinal cord. Within the graft white matter (WM) we identified astrocytes that form glial limitans, myelinating oligodendrocytes, and myelinated axons with paranodes. Within the graft grey matter (GM) we identified cholinergic, glutamatergic, serotonergic and dopaminergic neurons. Furthermore, we demonstrate the presence of ESC-derived complex vasculature that includes the presence of blood brain barrier. In addition to the formation of mature spinal cord tissue, we describe factors that drive this process. Specifically, we identify Flk1+ cells as necessary for spinal cord formation, and synaptic connectivity with the host spinal cord and formation of host-graft chimeric vasculature as contributing factors. This model can be used to study spinal cord organogenesis, and as an in vivo drug discovery platform for screening potential therapeutic compounds and their toxicity.
Collapse
|
17
|
Liu F, Kc P, Ni L, Zhang G, Zhe J. A microfluidic competitive immuno-aggregation assay for high sensitivity cell secretome detection. Organogenesis 2018; 14:67-81. [PMID: 29883244 DOI: 10.1080/15476278.2018.1461306] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We report a high-sensitivity cell secretome detection method using competitive immuno-aggregation and a micro-Coulter counter. A target cell secretome protein competes with anti-biotin-coated microparticles (MPs) to bind with a biotinylated antibody (Ab), causing decreased aggregation of the functionalized MPs and formation of a mixture of MPs and aggregates. In comparison, without the target cell secretome protein, more microparticles are functionalized, and more aggregates are formed. Thus, a decrease in the average volume of functionalized microparticles/aggregates indicates an increase in cell secretome concentration. This volume change is measured by the micro-Coulter counter, which is used to quantitatively estimate the cell secretome concentration. Vascular endothelial growth factor (VEGF), one of the key cell secretome proteins that regulate angiogenesis and vascular permeabilization, was used as the target protein to demonstrate the sensing principle. A standard calibration curve was generated by testing samples with various VEGF concentrations. A detection range from 0.01 ng/mL to 100.00 ng/mL was achieved. We further demonstrated the quantification of VEGF concentration in exogenous samples collected from the secretome of human mesenchymal stem cells (hMSCs) at different incubation times. The results from the assay agree well with the results of a parallel enzyme-linked immunoabsorbent assay (ELISA) test, indicating the specificity and reliability of the competitive immuno-aggregation assay. With its simple structure and easy sample preparation, this assay not only enables high sensitivity detection of VEGF but also can be readily extended to other types of cell secretome analysis as long as the specific Ab is known.
Collapse
Affiliation(s)
- Fan Liu
- a Department of Mechanical Engineering , The University of Akron , Akron , Ohio , United States
| | - Pawan Kc
- b Department of Biomedical Engineering , The University of Akron , Akron , Ohio , United States
| | - Liwei Ni
- a Department of Mechanical Engineering , The University of Akron , Akron , Ohio , United States
| | - Ge Zhang
- b Department of Biomedical Engineering , The University of Akron , Akron , Ohio , United States
| | - Jiang Zhe
- a Department of Mechanical Engineering , The University of Akron , Akron , Ohio , United States
| |
Collapse
|
18
|
Pombero A, Garcia-Lopez R, Estirado A, Martinez S. Vascular pattern of the dentate gyrus is regulated by neural progenitors. Brain Struct Funct 2018; 223:1971-1987. [PMID: 29306978 DOI: 10.1007/s00429-017-1603-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/28/2017] [Indexed: 01/19/2023]
Abstract
Neurogenesis is a vital process that begins during early embryonic development and continues until adulthood, though in the latter case, it is restricted to the subventricular zone and the subgranular zone of the dentate gyrus (DG). In particular, the DG's neurogenic properties are structurally and functionally unique, which may be related to its singular vascular pattern. Neurogenesis and angiogenesis share molecular signals and act synergistically, supporting the concept of a neurogenic niche as a functional unit between neural precursors cells and their environment, in which the blood vessels play an important role. Whereas it is well known that vascular development controls neural proliferation in the embryonary and in the adult brain, by releasing neurotrophic factors; the potential influence of neural cells on vascular components during angiogenesis is largely unknown. We have demonstrated that the reduction of neural progenitors leads to a significant impairment of vascular development. Since VEGF is a potential regulator in the neurogenesis-angiogenesis crosstalk, we were interested in assessing the possible role of this molecule in the hippocampal neurovascular development. Our results showed that VEGF is the molecule involved in the regulation of vascular development by neural progenitor cells in the DG.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Blood Vessels/physiology
- CD13 Antigens/metabolism
- Cell Differentiation
- Cell Proliferation
- Dentate Gyrus/anatomy & histology
- Dentate Gyrus/embryology
- Dentate Gyrus/growth & development
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Developmental/physiology
- Ki-67 Antigen/metabolism
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neovascularization, Physiologic/physiology
- Nerve Tissue Proteins/metabolism
- Nestin/genetics
- Nestin/metabolism
- Neural Stem Cells/physiology
- Neurogenesis/physiology
- RNA, Messenger
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
Collapse
Affiliation(s)
- Ana Pombero
- IMIB-Arrixaca, University of Murcia, Av. Teniente Flomesta, 5, 30003, Murcia, Spain
| | - Raquel Garcia-Lopez
- IMIB-Arrixaca, University of Murcia, Av. Teniente Flomesta, 5, 30003, Murcia, Spain
| | - Alicia Estirado
- IMIB-Arrixaca, University of Murcia, Av. Teniente Flomesta, 5, 30003, Murcia, Spain
| | - Salvador Martinez
- Instituto de Neurociencias, UMH-CSIC, Campus de San Juan, 03550, Alicante, Spain.
- Centro de Investigación Biomédica En Red en Salud Mental (CIBERSAM), Madrid, Spain.
| |
Collapse
|
19
|
Tang MM, Lin WJ, Zhang JT, Zhao YW, Li YC. Exogenous FGF2 reverses depressive-like behaviors and restores the suppressed FGF2-ERK1/2 signaling and the impaired hippocampal neurogenesis induced by neuroinflammation. Brain Behav Immun 2017; 66:322-331. [PMID: 28529071 DOI: 10.1016/j.bbi.2017.05.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/10/2017] [Accepted: 05/17/2017] [Indexed: 01/03/2023] Open
Abstract
Our previous work demonstrated that neuroinflammation evoked by triple repeated central LPS challenges inhibited adult hippocampal neurogenesis that were correlated with the depressive-like behavioral symptoms induced by neuroinflammation. These findings suggest that hippocampal neurogenesis might be one of biological mechanisms underlying depression induced by neuroinflammation and targeting neurogenesis might lead to new therapeutic strategies for the treatment of depression. In this study, we manipulated adult hippocampal neurogenesis using fibroblast growth factor 2 (FGF2), one crucial molecule modulating cell proliferation and survival in central nervous system, and investigate the involvement and the potential therapeutic effects of FGF2 on neuroinflammation-induced depression. Central lipopolysaccharides (LPS) challenges were used as previously to evoke the neuroinflammatory state in the brain of rat. Exogenous FGF2 was infused into lateral ventricle during the neuroinflammatory state. It was found that the protein expression of FGF2 in hippocampus was inhibited by neuroinflammation. The activation of extracellular signal-regulated kinase (ERK), the downstream molecule of FGF2, was also inhibited by neuroinflammation. Exogenous FGF2 infusions prevented the decrease in phosphorylation of ERK1/2 under neuroinflammation state. Exogenous FGF2 reversed depressive-like behaviors and the impaired hippocampal neurogenesis induced by neuroinflammation. These findings provide evidence that the FGF2-ERK1/2 pathway is involved in the pathophysiology of depressive-like behaviors, and manipulating the neurogenesis pathway is a viable therapeutic approach to inflammation-associated depression.
Collapse
Affiliation(s)
- Ming-Ming Tang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wen-Juan Lin
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China; Brain-Behavior Research Center, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Jun-Tao Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Wei Zhao
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying-Cong Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
| |
Collapse
|
20
|
Disrupting nNOS-PSD-95 coupling in the hippocampal dentate gyrus promotes extinction memory retrieval. Biochem Biophys Res Commun 2017; 493:862-868. [DOI: 10.1016/j.bbrc.2017.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 08/31/2017] [Accepted: 09/02/2017] [Indexed: 11/20/2022]
|
21
|
Gomez-Roman N, Stevenson K, Gilmour L, Hamilton G, Chalmers AJ. A novel 3D human glioblastoma cell culture system for modeling drug and radiation responses. Neuro Oncol 2017; 19:229-241. [PMID: 27576873 PMCID: PMC5463789 DOI: 10.1093/neuonc/now164] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common primary brain tumor, with dismal prognosis. The failure of drug-radiation combinations with promising preclinical data to translate into effective clinical treatments may relate to the use of simplified 2-dimensional in vitro GBM cultures. Methods We developed a customized 3D GBM culture system based on a polystyrene scaffold (Alvetex) that recapitulates key histological features of GBM and compared it with conventional 2D cultures with respect to their response to radiation and to molecular targeted agents for which clinical data are available. Results In 3 patient-derived GBM lines, no difference in radiation sensitivity was observed between 2D and 3D cultures, as measured by clonogenic survival. Three different molecular targeted agents, for which robust clinical data are available were evaluated in 2D and 3D conditions: (i) temozolomide, which improves overall survival and is standard of care for GBM, exhibited statistically significant effects on clonogenic survival in both patient-derived cell lines when evaluated in the 3D model compared with only one cell line in 2D cells; (ii) bevacizumab, which has been shown to increase progression-free survival when added to standard chemoradiation in phase III clinical trials, exhibited marked radiosensitizing activity in our 3D model but had no effect on 2D cells; and (iii) erlotinib, which had no efficacy in clinical trials, displayed no activity in our 3D GBM model, but radiosensitized 2D cells. Conclusions Our 3D model reliably predicted clinical efficacy, strongly supporting its clinical relevance and potential value in preclinical evaluation of drug-radiation combinations for GBM.
Collapse
Affiliation(s)
- Natividad Gomez-Roman
- Wolfson Wohl Translational Cancer Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Katrina Stevenson
- Wolfson Wohl Translational Cancer Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lesley Gilmour
- Wolfson Wohl Translational Cancer Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Graham Hamilton
- Wolfson Wohl Translational Cancer Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Anthony J Chalmers
- Wolfson Wohl Translational Cancer Research Center, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
22
|
Yi BR, Kim SU, Choi KC. Synergistic effect of therapeutic stem cells expressing cytosine deaminase and interferon-beta via apoptotic pathway in the metastatic mouse model of breast cancer. Oncotarget 2017; 7:5985-99. [PMID: 26716512 PMCID: PMC4868735 DOI: 10.18632/oncotarget.6719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/25/2015] [Indexed: 11/25/2022] Open
Abstract
As an approach to improve treatment of breast cancer metastasis to the brain, we employed genetically engineered stem cells (GESTECs, HB1.F3 cells) consisting of neural stem cells (NSCs) expressing cytosine deaminase and the interferon-beta genes, HB1.F3.CD and HB1.F3.CD.IFN-β. In this model, MDA-MB-231/Luc breast cancer cells were implanted in the right hemisphere of the mouse brain, while pre-stained GESTECs with redfluorescence were implanted in the contralateral brain. Two days after stem cells injection, 5-fluorocytosine (5-FC) was administrated via intraperitoneal injection. Histological analysis of extracted brain confirmed the therapeutic efficacy of GESTECs in the presence of 5-FC based on reductions in density and aggressive tendency of breast cancer cells, as well as pyknosis, karyorrhexis, and karyolysis relative to a negative control. Additionally, expression of PCNA decreased in the stem cells treated group. Treatment of breast cancer cells with 5-fluorouracil (5-FU) increased the expression of pro-apoptotic and anti-proliferative factor, BAX and p21 protein through phosphorylation of p53 and p38. Moreover, analysis of stem cell migratory ability revealed that MDA-MB-231 cells endogenously secreted VEGF, and stem cells expressed their receptor (VEGFR2). To confirm the role of VEGF/VEGFR2 signaling in tumor tropism of stem cells, samples were treated with the VEGFR2 inhibitor, KRN633. The number of migrated stem cells decreased significantly in response to KRN633 due to Erk1/2 activation and PI3K/Akt inhibition. Taken together, these results indicate that treatment with GESTECs, particularly HB1.F3.CD.IFN-β co-expressing CD.IFN-β, may be a useful strategy for treating breast cancer metastasis to the brain in the presence of a prodrug.
Collapse
Affiliation(s)
- Bo-Rim Yi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Seung U Kim
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.,TheraCell Bio and Science, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
23
|
Wang JZ, Fang Y, Ji WD, Xu H. LXR agonists promote the proliferation of neural progenitor cells through MEK-ERK pathway. Biochem Biophys Res Commun 2016; 483:216-222. [PMID: 28034754 DOI: 10.1016/j.bbrc.2016.12.163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 12/23/2016] [Indexed: 11/18/2022]
Abstract
The liver X receptors (LXRs) are transcriptional regulators of lipid homeostasis and may be critical for neurodegeneration and neurogenesis in vivo. However, it remains largely unknown about the role of LXRs and its agonists in the in vitro proliferation of neural progenitor cells (NPCs). Here we revealed for the first time that LXRs were markedly expressed in mouse NPCs and were critical for the in vitro proliferation. LXR agonists GW3965 and LXR623 promoted the proliferation of wildtype NPCs, but not NPCs from LXR double-knockout mice. Mechanistically, phosphorylation of MEK1/2 and ERK1/2 in NPCs was enhanced upon LXR agonist treatment, while abrogation of MEK/ERK phosphorylation by the inhibitors PD98059 and U0126 impaired the proliferation of wildtype NPCs in the presence or absence of LXR agonists. Collectively, our findings suggest that LXR agonists GW3965 and LXR623 can stimulate the NPC proliferation in LXR- and MEK/ERK-dependent manner.
Collapse
Affiliation(s)
- Jing-Zhong Wang
- Department of Neurology, The First People's Hospital of Shangqiu City, No 292, South Kaixuan Rd., Shangqiu 476100, Henan, People's Republic of China.
| | - Yan Fang
- Department of Neurology, The First People's Hospital of Shangqiu City, No 292, South Kaixuan Rd., Shangqiu 476100, Henan, People's Republic of China
| | - Wei-Dong Ji
- Department of Neurology, The First People's Hospital of Shangqiu City, No 292, South Kaixuan Rd., Shangqiu 476100, Henan, People's Republic of China
| | - Hui Xu
- Department of Neurology, The First People's Hospital of Shangqiu City, No 292, South Kaixuan Rd., Shangqiu 476100, Henan, People's Republic of China
| |
Collapse
|
24
|
Li H, Wang Y, Chen L, Han L, Li L, He H, Li Y, Huang N, Ren H, Pei F, Li G, Cheng J, Wang W. The role of MIF, cyclinD1 and ERK in the development of pulmonary hypertension in broilers. Avian Pathol 2016; 46:202-208. [PMID: 27706945 DOI: 10.1080/03079457.2016.1245409] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pulmonary hypertension (PH) is a major disease in the broiler breeding industry. During PH, the pulmonary artery undergoes remodelling, which is caused by pulmonary vascular smooth muscle cell proliferation. CyclinD1 regulates cell proliferation. This study investigated the role of cyclinD1 in the development of PH in broilers, and which bioactivators and signalling pathway are involved in the pathological process. The PH group contained 3-4-week-old broilers with clinical PH, and the healthy group broilers from the same flock without PH. Histopathology indicated pulmonary arterial walls were thicker in the PH group compared with the healthy group. Target gene expressions of macrophage migration inhibitory factor (MIF), extracellular signal-regulated kinase (ERK), and cyclinD1 detected by quantitative real-time PCR were upregulated in the PH group compared with the healthy group. Immunohistochemistry showed MIF, phosphorylated ERK (p-ERK) and cyclinD1 were present on pulmonary vascular walls; MIF was present in the cytoplasm of arterial endothelial cells and smooth muscle cells; p-ERK and cyclinD1 were present in smooth muscle cell cytoplasm. Western blotting demonstrated that MIF, p-ERKand cyclinD1 levels were significantly higher (P < 0.01) in the PH group compared with the healthy group. In summary, increased MIF in PH broiler pulmonary arteries upregulated cyclinD1 via the ERK signalling pathway to induce pulmonary vascular smooth muscle cell proliferation, causing pulmonary artery remodelling and hypertension.
Collapse
Affiliation(s)
- Haoyun Li
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Yanmei Wang
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Lingli Chen
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Lijuan Han
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Lifang Li
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Han He
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Yuan Li
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Nan Huang
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Hao Ren
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Fangying Pei
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Guilan Li
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Jia Cheng
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| | - Wenkui Wang
- a College of Animal Science and Technology , Shanxi Agricultural University , Taigu , People's Republic of China
| |
Collapse
|
25
|
Xiong A, Kundu S, Forsberg M, Xiong Y, Bergström T, Paavilainen T, Kjellén L, Li JP, Forsberg-Nilsson K. Heparanase confers a growth advantage to differentiating murine embryonic stem cells, and enhances oligodendrocyte formation. Matrix Biol 2016; 62:92-104. [PMID: 27890389 DOI: 10.1016/j.matbio.2016.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 01/23/2023]
Abstract
Heparan sulfate proteoglycans (HSPGs), ubiquitous components of mammalian cells, play important roles in development and homeostasis. These molecules are located primarily on the cell surface and in the pericellular matrix, where they interact with a multitude of macromolecules, including many growth factors. Manipulation of the enzymes involved in biosynthesis and modification of HSPG structures alters the properties of stem cells. Here, we focus on the involvement of heparanase (HPSE), the sole endo-glucuronidase capable of cleaving of HS, in differentiation of embryonic stem cells into the cells of the neural lineage. Embryonic stem (ES) cells overexpressing HPSE (Hpse-Tg) proliferated more rapidly than WT ES cells in culture and formed larger teratomas in vivo. In addition, differentiating Hpse-Tg ES cells also had a higher growth rate, and overexpression of HPSE in NSPCs enhanced Erk and Akt phosphorylation. Employing a two-step, monolayer differentiation, we observed an increase in HPSE as wild-type (WT) ES cells differentiated into neural stem and progenitor cells followed by down-regulation of HPSE as these NSPCs differentiated into mature cells of the neural lineage. Furthermore, NSPCs overexpressing HPSE gave rise to more oligodendrocytes than WT cultures, with a concomitant reduction in the number of neurons. Our present findings emphasize the importance of HS, in neural differentiation and suggest that by regulating the availability of growth factors and, or other macromolecules, HPSE promotes differentiation into oligodendrocytes.
Collapse
Affiliation(s)
- Anqi Xiong
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Soumi Kundu
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Maud Forsberg
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Yuyuan Xiong
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Tobias Bergström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Tanja Paavilainen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Lena Kjellén
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
26
|
Tumor Necrosis Factor Alpha Induces Neural Stem Cell Apoptosis Through Activating p38 MAPK Pathway. Neurochem Res 2016; 41:3052-3062. [PMID: 27528245 DOI: 10.1007/s11064-016-2024-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 07/20/2016] [Accepted: 07/29/2016] [Indexed: 12/21/2022]
Abstract
Tumor necrosis factor alpha (TNF-α) is an essential cytokine that mediates cell death and has been shown to play a potential role in inducing neural stem cell (NSC) apoptosis. We have previously shown that TNF-α antagonist etanercept can suppress the transplanted NSC apoptosis induced by TNF-α in spinal cord injury (SCI) sites; however, the precise molecular mechanism remains unclear. This study aimed to investigate the signaling pathways responsible for TNF-α-induced apoptosis in NSCs. TNF-α treatment impairs cell viability and increases apoptosis of NSCs in concentration- and time-dependent manners. This is embodied in an increase in Bax and cleaved caspase-3 production, coupled with decreased Bcl-2 levels. Additionally, TNF-α remarkably increased the expression of phosphatidylinositol p38 Mitogen-activated protein kinase (p38 MAPK) in NSCs. p38 MAPK regulates apoptosis, acting as an apoptotic signal due to TNF-α exposure. TNF-α-induced apoptosis was significantly alleviated by the p38 MAPK pathway inhibitor SB203580, as well as targeted inhibition of p38 gene in NSCs, or TNF-α antagonist etanercept. These results suggest that TNF-α induces NSCs apoptosis by activating the p38 MAPK signaling pathway and etanercept acts as an effective TNF-α antagonist to prevent p38 MAPK-dependent apoptosis induced by TNF-α in NSCs. Our research represents a potential gene targeting that can prevent unnecessary grafted cell death after transplantation into the SCI models.
Collapse
|
27
|
Belagodu AP, Zendeli L, Slater BJ, Galvez R. Blocking elevated VEGF‐A attenuates non‐vasculature Fragile X syndrome abnormalities. Dev Neurobiol 2016; 77:14-25. [DOI: 10.1002/dneu.22404] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/15/2016] [Accepted: 05/31/2016] [Indexed: 01/19/2023]
Affiliation(s)
- Amogh P. Belagodu
- Neuroscience Program, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| | - Liridon Zendeli
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| | - Bernard J. Slater
- Neuroscience Program, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| | - Roberto Galvez
- Neuroscience Program, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Psychology DepartmentUniversity of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| |
Collapse
|
28
|
Xue F, Liu L, Fan J, He S, Li R, Peng ZW, Wang BR. Interleukin-1β promotes the neurogenesis of carotid bodies by stimulating the activation of ERK1/2. Respir Physiol Neurobiol 2015; 219:78-84. [PMID: 26327233 DOI: 10.1016/j.resp.2015.08.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 01/31/2023]
Abstract
The carotid body (CB) is a complex sensory organ that functions to sense homeostatic O2 in the blood. Previous studies have shown that CBs express interleukin (IL)-1 receptor type I and that the chemosensitivity of CBs is increased following stimulation with pro-inflammatory cytokines. However, the effects of pro-inflammatory cytokines, such as IL-1β, on the neurogenesis of CB are unclear. Thus, in this study, we aimed to assess the effects of IL-1β and intermittent hypobaric hypoxia (IHH) plus IL-1β on the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, tyrosine hydroxylase (TH) and the expression of nestin, a well-established stem cell marker in the nervous system. The results showed that TH, nestin expression and ERK1/2 phosphorylation were increased in the rat CB following intraperitoneal injection of IL-1β. Moreover, IL-1β had additive effects on IHH. These results suggested that the plasticity of CB was increased following treatment with IL-1β and that ERK1/2 may be involved in neurogenic signaling in CBs.
Collapse
Affiliation(s)
- Fen Xue
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Ling Liu
- Institute of Neuroscience, Fourth Military Medical University, Xi'an 710032, China
| | - Juan Fan
- College of Life Science, Shaanxi Normal University, China
| | - ShanShan He
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Rui Li
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Zheng-Wu Peng
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China.
| | - Bai-Ren Wang
- Institute of Neuroscience, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
29
|
Kim KT, Kim HJ, Cho DC, Bae JS, Park SW. Substance P stimulates proliferation of spinal neural stem cells in spinal cord injury via the mitogen-activated protein kinase signaling pathway. Spine J 2015; 15:2055-65. [PMID: 25921821 DOI: 10.1016/j.spinee.2015.04.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/20/2015] [Accepted: 04/20/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Substance P (SP) is a neuropeptide that can influence neural stem/progenitor cell (NSPC) proliferation and neurogenesis in the brain. However, we could not find any experimental study that investigates SP action in the spinal cord. PURPOSE The aims of our study were to investigate the potential of the neuropeptide SP in promoting the proliferation of spinal cord-derived NSPCs (SC-NSPCs) after spinal cord injury (SCI) and to clarify the roles of the mitogen-activated protein (MAP) kinase signaling pathway in the process. STUDY DESIGN This is a randomized animal study. METHODS The SC-NSPCs were suspended in 100 μL of a neurobasal medium containing SP (binds neurokinin-1 receptor [NK1R]) or L-703,606 (NK1R antagonist) and cultured in a 96-well plate for 5 days. A cell proliferation assay was performed using a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. A cord clipping method was used for the SCI model. Substance P and the NK1R antagonist (L-703,606) were infused intrathecally in SCI and sham models. Neural stem/progenitor cell proliferation was evaluated with immunostaining for bromodeoxyuridine (BrdU) and the immature neural marker nestin. An immunoblotting method was used for evaluating the MAP kinase signaling protein that contains extracellular signal-regulated kinases (ERKs and p38) and β-actin as the control group. RESULTS In vitro, SP (0.01-10 μmol/L) increased the proliferation of cultured SC-NSPCs, with a peak increase of 35±2% at the 0.1 μmol/L concentration. Substance P of 0.1 μmol/L continuously increased SC-NSPC proliferation from 6 hours to 5 days, whereas the proliferation decreased from 18% to 98% with L-703,606 (1-10 μM). Intrathecal infusion of SP (1 μmol/L) for 7 days significantly increased the number of proliferating NPSCs (cells positive for both BrdU and nestin) in the spinal cord (by 120±17%, p<.05) in adult rats, but infusion of L-703,606 (10 μmol/L) significantly decreased the post-SCI induction of NPSC proliferation in the spinal cord (by 87±4%). Also, SP stimulates proliferation of SC-NSPCs via the MAP kinase signaling pathway, especially the phosphorylated ERK and phosphorylated p38 proteins. The phosphorylated ERK and phosphorylated p38 protein levels increased with SP (0.1 μmol/L, p<.05). CONCLUSIONS These data indicate that SP can promote proliferation of SC-NSPCs in SCI and normal conditions and have important roles in neuronal regeneration after SCI. Also, ERKs and p38 MAP kinases are important signaling proteins in this process.
Collapse
Affiliation(s)
- Kyoung-Tae Kim
- Department of Neurosurgery, Kyungpook National University Hospital, 50 Samduk-2-ga, Jung-gu, Daegu 700-721, Republic of Korea.
| | - Hye-Jeong Kim
- Department of Neurosurgery, Kyungpook National University Hospital, 50 Samduk-2-ga, Jung-gu, Daegu 700-721, Republic of Korea
| | - Dae-Chul Cho
- Department of Neurosurgery, Kyungpook National University Hospital, 50 Samduk-2-ga, Jung-gu, Daegu 700-721, Republic of Korea
| | - Jae-Sung Bae
- Department of Physiology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 700-842, Republic of Korea
| | - Seung-Won Park
- Department of Neurosurgery, College of Medicine, Chung-Ang University Hospital, 224-1 Heukseok dong, Dongjak-gu, Seoul 156-755, Republic of Korea
| |
Collapse
|
30
|
Yang D, Wang ZQ, Deng JQ, Liao JY, Wang X, Xie J, Deng MM, Lü MH. Adipose-derived stem cells: A candidate for liver regeneration. J Dig Dis 2015; 16:489-98. [PMID: 26121206 DOI: 10.1111/1751-2980.12268] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The scarcity of donor livers and the impracticality of hepatocyte transplantation represent the biggest obstacles for the treatment of liver failure. Adipose-derived stem cells, with their ability to differentiate into the hepatic lineage, provide a reliable alternative cell source with clear ethical and practical advantages. Moreover, adipose-derived stem cells can effectively repair liver damage by the dominant indirect pattern and increase the number of hepatocytes by the secondary direct pattern. In recent years, the development of the indirect pattern, which mainly includes immunomodulatory and trophic effects, has become a hot topic in the field of cell engineering. Therefore, adipose-derived stem cells are considered to be ideal therapeutic stem cells for human liver regeneration. In this article, we reviewed the advantages of adipose-derived stem cells in liver regeneration, and explore their underlying mechanisms.
Collapse
Affiliation(s)
- Dan Yang
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Zhong Qiong Wang
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Jia Qi Deng
- School of Foreign Languages of Sichuan Medical University, Luzhou, Sichuan Province, China
| | - Jing Yuan Liao
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Xuan Wang
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Jing Xie
- Department of Pediatric Surgery, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Ming Ming Deng
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Mu Han Lü
- Department of Gastroenterology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan Province, China
| |
Collapse
|
31
|
Chen YB, Lan YW, Hung TH, Chen LG, Choo KB, Cheng WTK, Lee HS, Chong KY. Mesenchymal stem cell-based HSP70 promoter-driven VEGFA induction by resveratrol promotes angiogenesis in a mouse model. Cell Stress Chaperones 2015; 20:643-52. [PMID: 25860916 PMCID: PMC4463926 DOI: 10.1007/s12192-015-0588-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/17/2015] [Accepted: 03/24/2015] [Indexed: 11/29/2022] Open
Abstract
Several studies of stem cell-based gene therapy have indicated that long-lasting regeneration following vessel ischemia may be stimulated through VEGFA gene therapy and/or MSC transplantation for reduction of ischemic injury in limb ischemia and heart failure. The therapeutic potential of MSC transplantation can be further improved by genetically modifying MSCs with genes which enhance angiogenesis following ischemic injury. In the present study, we aimed to develop an approach in MSC-based therapy for repair and mitigation of ischemic injury and regeneration of damaged tissues in ischemic disease. HSP70 promoter-driven VEGFA expression was induced by resveratrol (RSV) in MSCs, and in combination with known RSV biological functions, the protective effects of our approach were investigated by using ex vivo aortic ring coculture system and a 3D scaffolds in vivo model. Results of this investigation demonstrated that HSP promoter-driven VEGFA expression in MSC increased approximately 2-fold over the background VEGFA levels upon HSP70 promoter induction by RSV. Exposure of HUVEC cells to medium containing MSC in which VEGFA had been induced by cis-RSV enhanced tube formation in the treated HUVEC cells. RSV-treated MSC cells differentiated into endothelial-like phenotypes, exhibiting markedly elevated expression of endothelial cell markers. These MSCs also induced aortic ring sprouting, characteristic of neovascular formation from pre-existing vessels, and additionally promoted neovascularization at the MSC transplantation site in a mouse model. These observations support a hypothesis that VEGFA expression induced by cis-RSV acting on the HSP70 promoter in transplanted MSC augments the angiogenic effects of stem cell gene therapy. The use of an inducible system also vastly reduces possible clinical risks associated with constitutive VEGFA expression.
Collapse
Affiliation(s)
- Young-Bin Chen
- />Institute of Biotechnology, National Taiwan University, Taipei, Taiwan Republic of China
| | - Ying-Wei Lan
- />Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan Republic of China
| | - Tsai-Hsien Hung
- />Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan Republic of China
| | - Lih-Geeng Chen
- />Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi, Taiwan Republic of China
| | - Kong-Bung Choo
- />Department of Preclinical Sciences, Faculty of Medicine and Health Sciences and Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Winston TK Cheng
- />Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan Republic of China
| | - Hsuan-Shu Lee
- />Institute of Biotechnology, National Taiwan University, Taipei, Taiwan Republic of China
- />Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan Republic of China
| | - Kowit-Yu Chong
- />Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan Republic of China
- />Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan Republic of China
- />Molecular Medicine Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan Republic of China
- />Department of Family Medicine, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan Republic of China
| |
Collapse
|
32
|
Improvement of the survival of human autologous fat transplantation by adipose-derived stem-cells-assisted lipotransfer combined with bFGF. ScientificWorldJournal 2015; 2015:968057. [PMID: 25695105 PMCID: PMC4324956 DOI: 10.1155/2015/968057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/01/2015] [Accepted: 01/04/2015] [Indexed: 12/22/2022] Open
Abstract
Adipose-derived stem cells (ASCs) transplanted along with autologous adipose tissue may improve fat graft survival; however, the efficacy of ASCs has been diluted by low vascularization. This study was designed to test the hypothesis that basic fibroblast growth factor (bFGF) may improve the effects of ASCs because it owns the property to boost angiogenesis. In the present study, human fat tissues were mixed with ASCs, ASCs plus 100 U bFGF, or medium as the control and then injected subcutaneously into immunologically compromised nude mice for 12 weeks. Our findings demonstrated that mixture with the ASCs significantly increased the weight and volume of the fat grafts compared to control grafts, and histological analysis revealed that both ASCs and ASCs plus bFGF grafts consisted predominantly of adipose tissue and had significantly less fibrosis but greater microvascular density compared with control and also grafts mixed with ASCs had a high expression of angiogenic factors. More importantly, the bFGF treated fat grafts shown elevate in survival, vascularization, and angiogenic factors expression when compared with the grafts that received ASCs alone. These results indicated that bFGF together with ASCs can enhance the efficacy of autologous fat transplantation and increase blood vessel generation involved in the benefits from bFGF.
Collapse
|
33
|
Karelina K, Liu Y, Alzate-Correa D, Wheaton KL, Hoyt KR, Arthur JSC, Obrietan K. Mitogen and stress-activated kinases 1/2 regulate ischemia-induced hippocampal progenitor cell proliferation and neurogenesis. Neuroscience 2014; 285:292-302. [PMID: 25451279 DOI: 10.1016/j.neuroscience.2014.10.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 09/30/2014] [Accepted: 10/20/2014] [Indexed: 01/10/2023]
Abstract
Pathophysiological conditions such as cerebral ischemia trigger the production of new neurons from the neurogenic niche within the subgranular zone (SGZ) of the dentate gyrus. The functional significance of ischemia-induced neurogenesis is believed to be the regeneration of lost cells, thus contributing to post-ischemia recovery. However, the cell signaling mechanisms by which this process is regulated are still under investigation. Here, we investigated the role of mitogen and stress-activated protein kinases (MSK1/2) in the regulation of progenitor cell proliferation and neurogenesis after cerebral ischemia. Using the endothelin-1 model of ischemia, wild-type (WT) and MSK1(-/-)/MSK2(-/-) (MSK dKO) mice were injected with BrdU and sacrificed 2 days, 4 weeks, or 6 weeks later for the analysis of progenitor cell proliferation, neurogenesis, and neuronal morphology, respectively. We report a decrease in SGZ progenitor cell proliferation in MSK dKO mice compared to WT mice. Moreover, MSK dKO mice exhibited reduced neurogenesis and a delayed maturation of ischemia-induced newborn neurons. Further, structural analysis of neuronal arborization revealed reduced branching complexity in MSK dKO compared to WT mice. Taken together, this dataset suggests that MSK1/2 plays a significant role in the regulation of ischemia-induced progenitor cell proliferation and neurogenesis. Ultimately, revealing the cell signaling mechanisms that promote neuronal recovery will lead to novel pharmacological approaches for the treatment of neurodegenerative diseases such as cerebral ischemia.
Collapse
Affiliation(s)
- K Karelina
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Y Liu
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - D Alzate-Correa
- Division of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - K L Wheaton
- Division of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - K R Hoyt
- Division of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - J S C Arthur
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - K Obrietan
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
34
|
Shin Y, Yang K, Han S, Park HJ, Seok Heo Y, Cho SW, Chung S. Reconstituting vascular microenvironment of neural stem cell niche in three-dimensional extracellular matrix. Adv Healthc Mater 2014; 3:1457-64. [PMID: 24523050 DOI: 10.1002/adhm.201300569] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/21/2013] [Indexed: 11/07/2022]
Abstract
Neural stem cells (NSCs) reside in a vascular microenvironment termed the "NSC niche." Blood vessels in the NSC niche play an important role in maintaining an appropriate balance between NSC self-renewal and differentiation that serves to maintain homeostasis. Understanding the role of brain vessels in the NSC niche will facilitate basic research in neurogenesis and vasculogenesis as well as aid the development of potential therapies for degenerative disorders. Here, an in vitro-reconstituted NSC-vascular niche consisting of a 3D brain vasculature and extracellular matrix (ECM) microenvironment that allows NSCs to adopt physiologically relevant phenotypes through the combined effects of ECM components, chemical gradients, and signaling effectors from the brain vasculature is described. The reconstituted niche can provide precise spatiotemporal control of the NSC niche, regulating self-renewal, proliferation and colony formation of NSCs, and suppressing neuronal generation but promoting NSC differentiation into astrocytes and oligodendrocytes. It is proved that Notch effectors regulate both the astrocyte differentiation and NSC self-renewal, but the astrocyte differentiation is more active in NSCs in close proximity to brain vasculature. A potential role of the other vascular microenvironmental factor of pigment epithelium-derived factor from brain vasculature in the regulation of NSC self-renewal is also proved.
Collapse
Affiliation(s)
- Yoojin Shin
- Department of Mechanical Engineering, Korea University, #512B, Innovation Hall, Anam, Seongbuk, Seoul, 136-713, South Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
Baptista S, Lasgi C, Benstaali C, Milhazes N, Borges F, Fontes-Ribeiro C, Agasse F, Silva AP. Methamphetamine decreases dentate gyrus stem cell self-renewal and shifts the differentiation towards neuronal fate. Stem Cell Res 2014; 13:329-41. [DOI: 10.1016/j.scr.2014.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 07/16/2014] [Accepted: 08/05/2014] [Indexed: 01/21/2023] Open
|
36
|
Koyama Y, Hayashi M, Nagae R, Tokuyama S, Konishi T. Endothelin-1 increases the expression of VEGF-R1/Flt-1 receptors in rat cultured astrocytes through ETB receptors. J Neurochem 2014; 130:759-69. [PMID: 24862165 DOI: 10.1111/jnc.12770] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 01/04/2023]
Abstract
Expressions of vascular endothelial growth factor (VEGF) receptors in astrocytes are increased in damaged brains. To clarify the regulatory mechanisms of VEGF receptors, the effects of endothelin-1 (ET-1) were examined in rat cultured astrocytes. Expressions of VEGF-R1 and -R2 receptor mRNA were at similar levels, whereas the mRNA expressions of VEGF-R3 and Tie-2, a receptor for angiopoietins, were lower. Placenta growth factor, a selective agonist of the VEGF-R1 receptor, induced phosphorylation of focal adhesion kinase (FAK) and extracellular signal regulated kinase 1/2 (ERK1/2). Phosphorylations of FAK and ERK 1/2 were also stimulated by VEGF-E, a selective VEGF-R2 agonist. Increased phosphorylations of FAK and ERK1/2 by VEGF165 were reduced by selective antagonists for VEGF-R1 and -R2. Treatment with ET-1 increased VEGF-R1 mRNA and protein levels. The effects of ET-1 on VEGF-R1 mRNA were mimicked by Ala(1,3,11,15) -ET-1, a selective agonist for ETB receptors, and inhibited by BQ788, an ETB antagonist. ET-1 did not affect the mRNA levels of VEGF-R2, -R3, and Tie-2. Pre-treatment with ET-1 potentiated the effects of placenta growth factor on phosphorylations of FAK and ERK1/2. These findings suggest that ET-1 induces up-regulation of VEGF-R1 receptors in astrocytes, and potentiates VEGF signals in damaged nerve tissues. To clarify the regulatory mechanisms of vascular endothelial growth factor (VEGF) receptors, the effects of endothelin-1 (ET-1) were examined in rat cultured astrocytes. Effects of selective VEGF-R1 and R2 agonist showed that these receptors were linked to focal adhesion kinase (FAK) and extracellular signal regulated kinase 1/2 (ERK1/2). Treatment with ET-1 increased expression of VEGF-R1, which was mediated by ETB receptors. Pre-treatment with ET-1 potentiated the VEGF-R1-mediated activations of FAK and ERK1/2. These findings suggest that ET-1 induces up-regulation of VEGF-R1 receptors in astrocytes.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | | | | | | | | |
Collapse
|
37
|
Zhang E, Shen J, So KF. Chinese traditional medicine and adult neurogenesis in the hippocampus. J Tradit Complement Med 2014; 4:77-81. [PMID: 24860729 PMCID: PMC4003705 DOI: 10.4103/2225-4110.130372] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adult neurogenesis is an important therapeutic target in treating neurological disorders. Adult neurogenesis takes place in two regions of the brain: Subventricular zone and dentate gyrus in the hippocampus. The progressive understanding on hippocampal neurogenesis in aging and mood disorders increases the demand to explore powerful and subtle interventions on hippocampal neurogenesis. Traditional Chinese herbal medicine provides an abundant pharmaceutical platform for modulating hippocampal neurogenesis. Recent progress in exploring the effects of Chinese herbal medicine and the related mechanisms opens a new direction for regeneration therapy. The current review gives a thorough summary of the research progress made in traditional Chinese herbal formulas, and the effective compounds in Chinese herbs which are beneficial on hippocampal neurogenesis and the possible mechanisms involved.
Collapse
Affiliation(s)
- Endong Zhang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China. ; Department of Anatomy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jiangang Shen
- School of Chinese Medicine, Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Kwok Fai So
- Department of Anatomy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China. ; Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China. ; The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China. ; GHM Institute of CNS Regeneration, and Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China
| |
Collapse
|
38
|
Nie H, Peng Z, Lao N, Wang H, Chen Y, Fang Z, Hou W, Gao F, Li X, Xiong L, Tan Q. Rosmarinic acid ameliorates PTSD-like symptoms in a rat model and promotes cell proliferation in the hippocampus. Prog Neuropsychopharmacol Biol Psychiatry 2014; 51:16-22. [PMID: 24418162 DOI: 10.1016/j.pnpbp.2014.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 01/03/2014] [Accepted: 01/03/2014] [Indexed: 01/14/2023]
Abstract
Rosmarinic acid (RA) is an important component of Chinese herbal medicine treatments and has been demonstrated to exert therapeutic effects in mood disorders. The present study was designed to assess the effects of RA on post-traumatic stress disorder (PTSD)-like symptoms, hippocampal cell proliferation and phosphorylation extracellular regulated protein kinases (pERK1/2) expression. We found that administration of RA (10mg/kg) alleviated PTSD-like symptoms in rats exposed to an enhanced single prolonged stress (ESPS) paradigm and restored hippocampal proliferation and pERK1/2 expression. Interestingly, the effects of RA were inhibited by the blockage of the ERK signaling. These data support the use of RA for treating PTSD and indicate that the ERK1/2 signaling cascade may play a critical role in the therapeutic efficacy of RA in treating such conditions.
Collapse
Affiliation(s)
- Huang Nie
- Department of Psychosomatic Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China; Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Zhengwu Peng
- Department of Psychosomatic Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China; Department of Toxicology, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Ning Lao
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Huaning Wang
- Department of Psychosomatic Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Yihuan Chen
- Department of Psychosomatic Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Zongping Fang
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Wugang Hou
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Fang Gao
- Institute of Neuroscience, The Fourth Military Medical University, Xi'an 710032, China
| | - Xia Li
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Lize Xiong
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| | - Qingrong Tan
- Department of Psychosomatic Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
39
|
Jang HJ, Kim JS, Choi HW, Jeon I, Choi S, Kim MJ, Song J, Do JT. Neural stem cells derived from epiblast stem cells display distinctive properties. Stem Cell Res 2014; 12:506-16. [PMID: 24463498 DOI: 10.1016/j.scr.2013.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 02/05/2023] Open
Abstract
Pluripotent stem cells can be derived from preimplantation and postimplantation mouse embryos. Embryonic stem cells (ESCs) derived from blastocysts are in a "naive" pluripotent state and meet all of the criteria for pluripotency, including the ability to generate live pups through tetraploid complementation. Epiblast stem cells (EpiSCs) derived from postimplantation epiblasts are in a "primed" pluripotent state. ESCs and EpiSCs show different phenotypes and gene expression patterns, and EpiSCs are thought to be less pluripotent than ESCs. In this study, we addressed whether EpiSCs can be differentiated into specialized cell types in vitro. To do this, we first derived EpiSCs from E5.5-6.5 mouse embryos containing the Oct4-GFP transgene. We found that EpiSCs expressed pluripotency markers and differentiated into all three germ layers in intro and in vivo. Interestingly, EpiSCs also efficiently differentiated into a homogenous population of neural stem cells (NSCs) in vitro. The EpiSC-derived NSCs (EpiSC-NSCs) expressed NSC markers (Nestin, Sox2, and Musashi), self-renewed for more than 20 passages, and differentiated into neuronal and glial neural cell subtypes in vitro. We then transplanted the EpiSC-NSCs into the neonatal mouse brains, and found that they were able to survive and differentiate into robust neurons and glial cells in the mouse brains, demonstrating that primed pluripotent EpiSCs efficiently form functional NSCs. We compared the global gene expression patterns of NSCs differentiated from EpiSC-NSCs, ESCs, and brain tissue and found that the expression patterns of most genes, including pluripotency and NSC specificity, were similarly clustered, but that the developmental process-related genes were distantly clustered. Moreover, the global gene expression pattern of brain-derived NSCs was more similar to that of ESC-derived NSCs than that of EpiSC-derived NSCs. Taken together, these results indicate that although NSCs, regardless of their origins, display very similar in vitro and in vivo differentiation properties, their global gene expression profiles may differ, depending on the pluripotency state, i.e., naive or primed.
Collapse
Affiliation(s)
- Hyo Jin Jang
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Jong Soo Kim
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hyun Woo Choi
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Iksoo Jeon
- Department of Biomedical Science, CHA University, 606-16 Yeoksam 1-dong, Kangnam-gu, Seoul 135-081, Republic of Korea
| | - Sol Choi
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Min Jung Kim
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Jihwan Song
- Department of Biomedical Science, CHA University, 606-16 Yeoksam 1-dong, Kangnam-gu, Seoul 135-081, Republic of Korea
| | - Jeong Tae Do
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea.
| |
Collapse
|
40
|
Sphingosine-1-phosphate-induced Flk-1 transactivation stimulates mouse embryonic stem cell proliferation through S1P1/S1P3-dependent β-arrestin/c-Src pathways. Stem Cell Res 2014; 12:69-85. [DOI: 10.1016/j.scr.2013.08.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 08/08/2013] [Accepted: 08/29/2013] [Indexed: 01/21/2023] Open
|
41
|
Liu D, Wang Z, Zhan J, Zhang Q, Wang J, Zhang Q, Xian X, Luan Q, Hao A. Hydrogen sulfide promotes proliferation and neuronal differentiation of neural stem cells and protects hypoxia-induced decrease in hippocampal neurogenesis. Pharmacol Biochem Behav 2013; 116:55-63. [PMID: 24246910 DOI: 10.1016/j.pbb.2013.11.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 10/24/2013] [Accepted: 11/07/2013] [Indexed: 12/27/2022]
Abstract
Accumulating evidence has suggested that hydrogen sulfide (H2S) acts as a novel neuro-modulator and neuroprotective agent; however, it remains to be investigated whether H2S has a direct effect on neural stem cells (NSCs). In the present study, we examined the effects of H2S donor, sodium hydrosulfide (NaHS) on mouse NSCs and hippocampal neurogenesis. We report here that NaHS promoted proliferation and neuronal differentiation of NSCs. Further analysis revealed that NaHS-induced proliferation was associated with phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 and neuronal differentiation was linked to altered expression of differentiation-related genes. In addition, C57BL/6 mice (1 day old) subjected to hypoxia were treated with NaHS to explore whether H2S would influence the neurogenesis of hippocampus. BrdU incorporation assay results showed that administration of NaHS could increase the number of proliferating cells in the dentate gyrus of hippocampus in the mice after hypoxia. Moreover, Morris water maze test showed that treatment with NaHS improved cognitive impairment after hypoxia in mice. These findings suggest that H2S may afford a novel therapeutic strategy to intervene in the progression of brain diseases.
Collapse
Affiliation(s)
- Dexiang Liu
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China; Institute of Medical Psychology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Zhen Wang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China; Institute of Physiology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Jingmin Zhan
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Qun Zhang
- Institute of Physiology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Jianmei Wang
- Institute of Physiology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Qingrui Zhang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Xiuying Xian
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Qinsong Luan
- Institute of Physiology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Aijun Hao
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, 44#, Wenhua Xi Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
42
|
Chen G, Shi X, Sun C, Li M, Zhou Q, Zhang C, Huang J, Qiu Y, Wen X, Zhang Y, Zhang Y, Yang S, Lu L, Zhang J, Yuan Q, Lu J, Xu G, Xue Y, Jin Z, Jiang C, Ying M, Liu X. VEGF-mediated proliferation of human adipose tissue-derived stem cells. PLoS One 2013; 8:e73673. [PMID: 24098328 PMCID: PMC3789739 DOI: 10.1371/journal.pone.0073673] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 07/21/2013] [Indexed: 12/21/2022] Open
Abstract
Human adipose tissue-derived stem cells (ADSCs) are an attractive multipotent stem cell source with therapeutic applicability across diverse fields for the repair and regeneration of acute and chronically damaged tissues. In recent years, there has been increasing interest in ADSC for tissue engineering applications. However, the mechanisms underlying the regulation of ADSC proliferation are not fully understood. Here we show that 47 transcripts are up-regulated while 23 are down-regulated in ADSC compared to terminally differentiated cells based on global mRNA profiling and microRNA profiling. Among the up-regulated genes, the expression of vascular endothelial growth factor (VEGF) is fine-tuned by miR-199a-5p. Further investigation indicates that VEGF accelerates ADSC proliferation whereas the multipotency of ADSC remains stable in terms of adipogenic, chondrogenic and osteogenic potentials after VEGF treatment, suggesting that VEGF may serve as an excellent supplement for accelerating ADSC proliferation during in vitro expansion.
Collapse
Affiliation(s)
- Guangfeng Chen
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiujuan Shi
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Sun
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Li
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Qing Zhou
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chen Zhang
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Huang
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Qiu
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangyi Wen
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Zhang
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yushan Zhang
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuzhang Yang
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lixia Lu
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jieping Zhang
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qionglan Yuan
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianwei Lu
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guotong Xu
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yunyun Xue
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zibing Jin
- Division of Ophthalmic Genetics, The Eye Hospital of Wenzhou Medical College and Lab for Stem Cell & Retinal Regeneration, School of Ophthalmology & Optometry, Wenzhou Medical College, Wenzhou, China
| | - Cizhong Jiang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- * E-mail: (XL); (CJ); (MY)
| | - Ming Ying
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences, Shenzhen University, Shenzhen, China
- * E-mail: (XL); (CJ); (MY)
| | - Xiaoqing Liu
- Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences, Shenzhen University, Shenzhen, China
- * E-mail: (XL); (CJ); (MY)
| |
Collapse
|
43
|
Nie H, Peng Z, Lao N, Dong H, Xiong L. Effects of sevoflurane on self-renewal capacity and differentiation of cultured neural stem cells. Neurochem Res 2013; 38:1758-1767. [PMID: 23756731 DOI: 10.1007/s11064-013-1074-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 05/02/2013] [Accepted: 05/09/2013] [Indexed: 12/11/2022]
Abstract
Sevoflurane anesthesia in infant rats can result in long-term cognitive impairment, possibly by inhibiting neurogenesis. The hippocampus is critical for memory consolidation and is one of only two mammalian brain regions where neural stem cells (NSCs) are renewed continuously throughout life. To elucidate the pathogenesis of sevoflurane-induced cognitive dysfunction, we measured the effects of clinical sevoflurane doses on the survival, proliferation, and differentiation of hippocampal NSCs. Neural stem cells were isolated from Sprague-Dawley rat embryos, expanded in vitro, and exposed to sevoflurane at 0.5, 1, or 1.5 minimal alveolar concentration (MAC) for 1 or 6 h. Two days after treatment, cell viability, cytotoxicity, and apoptosis rate were estimated by WST-1 assay, lactate dehydrogenase (LDH) activity, and TdT-mediated dUTP-biotin nick end labeling (TUNEL), respectively, while proliferation rate was assessed by 5-ethynyl-2'-deoxyuridine (BrdU) incorporation and Ki67 staining. Differentiation was assayed 7 days after treatment by immunocytochemistry and Western blots of neuron and glial markers. The phosphorylation level of p44/42 extracellular regulated kinases (ERK1/2) was measured in the proliferation and differentiation phases respectively. Sevoflurane at 1 MAC or 1.5 MAC for 1 h increased viable cell number whereas a 6 h exposure at these same concentrations suppressed proliferation and promoted apoptotic death (P < 0.01). Sevoflurane had no effect on NSC differentiation, and a sub-clinical concentration (0.5 MAC) altered neither proliferation nor viability. The phosphorylation level of ERK1/2 increased after 1 h of 1 MAC or 1.5 MAC of sevoflurane exposure in the proliferation phase, but not in the differentiation phase. Brief (1 h) exposure to sevoflurane at clinical concentrations enhanced proliferation of cultured NSCs possibly mediated by ERK1/2, but a 6 h exposure suppressed proliferation and induced apoptosis. Prolonged sevoflurane exposure may decrease the self-renewal capacity of hippocampal NSCs, resulting in cognitive deficits.
Collapse
Affiliation(s)
- Huang Nie
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | | | | | | | | |
Collapse
|
44
|
Gao JH, Wen SL, Yang WJ, Lu YY, Tong H, Huang ZY, Liu ZX, Tang CW. Celecoxib ameliorates portal hypertension of the cirrhotic rats through the dual inhibitory effects on the intrahepatic fibrosis and angiogenesis. PLoS One 2013; 8:e69309. [PMID: 23922700 PMCID: PMC3724827 DOI: 10.1371/journal.pone.0069309] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 06/12/2013] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Increased intra-hepatic resistance to portal blood flow is the primary factor leading to portal hypertension in cirrhosis. Up-regulated expression of cyclooxygenase-2 (COX-2) in the cirrhotic liver might be a potential target to ameliorate portal hypertension. OBJECTIVE To verify the effect of celecoxib, a selective inhibitor of COX-2, on portal hypertension and the mechanisms behind it. METHODS Cirrhotic liver model of rat was established by peritoneal injection of thiacetamide (TAA). 36 rats were randomly assigned to control, TAA and TAA+celecoxib groups. Portal pressures were measured by introduction of catheters into portal vein. Hepatic fibrosis was assessed by the visible hepatic fibrotic areas and mRNAs for collagen III and α-SMA. The neovasculature was determined by hepatic vascular areas, vascular casts and CD31 expression. Expressions of COX-2, vascular endothelial growth factor (VEGF), VEGF receptor-2 (VEGFR-2) and related signal molecules were quantitated. RESULTS Compared with TAA group, the portal pressure in TAA+celecoxib group was significantly decreased by 17.8%, p<0.01. Celecoxib treatment greatly reduced the tortuous hepatic portal venules. The data of fibrotic areas, CD31expression, mRNA levels of α-SMA and collagen III in TAA+celecoxib group were much lower than those in TAA group, p<0.01. Furthermore, the up-regulation of hepatic mRNA and protein levels of VEGF, VEGFR-2 and COX-2 induced by TAA was significantly inhibited after celecoxib treatment. The expressions of prostaglandin E2 (PGE2), phosphorylated extracellular signal-regulated kinase (p-ERK), hypoxia-inducible factor-1α (HIF-1α), and c-fos were also down-regulated after celecoxib treatment. CONCLUSIONS Long term administration of celecoxib can efficiently ameliorate portal hypertension in TAA rat model by its dual inhibitory effects on the intrahepatic fibrosis and angiogenesis. The anti-angiogenesis effect afforded by celecoxib may attribute to its modulation on VEGF/VEGFR-2 through the down-regulation of integrated signal pathways involving PGE2- HIF-1α- VEGF and p-ERK- c-fos- VEGFR-2.
Collapse
Affiliation(s)
- Jin-Hang Gao
- Division of Peptides Related with Human Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Shi-Lei Wen
- Division of Peptides Related with Human Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen-Juan Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yao-Yao Lu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Tong
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi-Yin Huang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang-Xu Liu
- Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Cheng-Wei Tang
- Division of Peptides Related with Human Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- * E-mail:
| |
Collapse
|
45
|
Peng Z, Zhang R, Wang H, Chen Y, Xue F, Wang L, Yang F, Chen Y, Liu L, Kuang F, Tan Q. Ziprasidone ameliorates anxiety-like behaviors in a rat model of PTSD and up-regulates neurogenesis in the hippocampus and hippocampus-derived neural stem cells. Behav Brain Res 2013; 244:1-8. [PMID: 23384713 DOI: 10.1016/j.bbr.2013.01.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 01/22/2013] [Accepted: 01/27/2013] [Indexed: 01/12/2023]
Abstract
Ziprasidone, a widely used atypical antipsychotic drug, has been demonstrated to have therapeutic effects in patients with post-traumatic stress disorder (PTSD), but its underlying mechanisms remain poorly understood. One possible explanation is that the neuroprotective and neurogenetic actions of ziprasidone can attenuate the neuronal apoptosis which occurs in the hippocampus. To test this hypothesis, the present study was designed to assess the effects of ziprasidone treatment on anxiety-like behaviors, hippocampal neurogenesis, and in vivo/in vitro expression of pERK1/2 and Bcl-2 in male Sprague-Dawley rats. The methodology involved 3 different experiments, and the investigations also included the assessment of U0126 interference in ziprasidone treatment. It was found that the in vivo, administration of ziprasidone not only reversed the anxiety-like behaviors in rats that exposed to an enhanced single prolonged stress paradigm, but also restored the proliferation and the protein expression of pERK1/2 and Bcl-2 in the hippocampus of these rats. Also, mild concentrations of ziprasidone promoted the in vitro proliferation of hippocampal-derived neural stem cells (NSCs) and increased the levels of pERK1/2 and Bcl-2 in NSCs. Interestingly, the observed effects of ziprasidone were inhibited by U0126. These data support the use of ziprasidone for the treatment of PTSD and indicate that the changes in the ERK1/2 signaling cascade may play a critical role in the pathophysiology of PTSD and its treatment modalities. Further investigations are needed to elucidate the detailed signal cascades involved in the pathophysiology of stress-related disorders, and confirm the efficacy of ziprasidone in anti-PTSD treatment.
Collapse
Affiliation(s)
- Zhengwu Peng
- Department of Psychosomatic Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Carmeliet P, Ruiz de Almodovar C, Carmen RDA. VEGF ligands and receptors: implications in neurodevelopment and neurodegeneration. Cell Mol Life Sci 2013; 70:1763-78. [PMID: 23475071 PMCID: PMC11113464 DOI: 10.1007/s00018-013-1283-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/15/2022]
Abstract
Intensive research in the last decade shows that the prototypic angiogenic factor vascular endothelial growth factor (VEGF) can have direct effects in neurons and modulate processes such as neuronal migration, axon outgrowth, axon guidance and neuronal survival. Depending on the neuronal cell type and the process, VEGF seems to exert these effects by signaling via different receptors. It is also becoming clear that other VEGF ligands such as VEGF-B, -C and -D can act in various neuronal cell types as well. Moreover, apart from playing a role in physiological conditions, VEGF and VEGF-B have been related to different neurological disorders. We give an update on how VEGF controls different processes during neurodevelopment as well as on its role in several neurodegenerative disorders. We also discuss recent findings demonstrating that other VEGF ligands influence processes such as neurogenesis and dendrite arborization and participate in neurodegeneration.
Collapse
Affiliation(s)
- Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, K.U.Leuven, 3000, Leuven, Belgium.
| | | | | |
Collapse
|
47
|
Okabe N, Nakamura E, Himi N, Narita K, Tsukamoto I, Maruyama T, Sakakibara N, Nakamura T, Itano T, Miyamoto O. Delayed administration of the nucleic acid analog 2Cl-C.OXT-A attenuates brain damage and enhances functional recovery after ischemic stroke. Brain Res 2013; 1506:115-31. [DOI: 10.1016/j.brainres.2013.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 02/06/2013] [Accepted: 02/06/2013] [Indexed: 01/28/2023]
|
48
|
Huperzine A promotes hippocampal neurogenesis in vitro and in vivo. Brain Res 2013; 1506:35-43. [DOI: 10.1016/j.brainres.2013.02.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 02/02/2013] [Accepted: 02/15/2013] [Indexed: 01/01/2023]
|
49
|
Peng ZW, Xue F, Wang HN, Zhang RG, Chen YC, Wang Y, Zhang LY, Fan J, Tan QR. Paroxetine up-regulates neurogenesis in hippocampus-derived neural stem cell from fetal rats. Mol Cell Biochem 2013; 375:105-113. [PMID: 23291919 DOI: 10.1007/s11010-012-1533-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 11/23/2012] [Indexed: 01/17/2023]
Abstract
Paroxetine is a widely used antidepressant in clinic. Besides its role in inhibition of serotonin reuptake, resent studies indicate that the increase of hippocampal neurogenesis is also involved in its pharmacology. However, only limited data are available in this regard and its effect on the hippocampus-derived neural stem cell (NSCs) has not been well elucidated. In present study, we utilized hippocampus-derived NSCs from fetal rats to investigate the direct effect of paroxetine on the neurogenesis of NSCs and explore the possible cellular and molecular mechanisms. The results showed that paroxetine not only promoted the proliferation of NSCs, but also promoted NSCs to differentiate into neurons other than glial cells. In addition, the elevated protein levels of phosphorylated ERK1/2, Bcl-2, and brain-derived neurotrophic factor were also observed after paroxetine was administered. Furthermore, the proliferative effect and promotion of NSCs differentiating predominantly into neurons of paroxetine was inhibited by U0126, an ERK1/2 phosphorylation inhibitor. In conclusion, these data indicate that paroxetine can promote neurogenesis of neural stem cells, and this effect might be mediated by ERK1/2 signal pathways.
Collapse
Affiliation(s)
- Zheng-Wu Peng
- Department of Psychosomatic Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
VEGF promotes proliferation of human glioblastoma multiforme stem-like cells through VEGF receptor 2. ScientificWorldJournal 2013; 2013:417413. [PMID: 23533349 PMCID: PMC3603324 DOI: 10.1155/2013/417413] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 02/03/2013] [Indexed: 12/24/2022] Open
Abstract
Cancer stem-like cells, which have been described as tumor-initiating cells or tumor-propagating cells, play a crucial role in our fundamental understanding of glioblastoma multiforme (GBM) and its recurrence. GBM is a lethal cancer, characterized by florid vascularization and aberrantly elevated vascular endothelial growth factor (VEGF). VEGF promotes tumorigenesis and angiogenesis of human GBM stem-like cells (GBSCs). However, whether and how VEGF contributes to GBSCs proliferation remain largely uncertain. In this study, human GBSCs were isolated from surgical specimens of glioblastoma and cultured in medium favored for stem cell growth. Neural Colony-Forming Cell Assay and ATP assay were performed to measure GBSC proliferation under normoxia (20% O2) and hypoxia (1% O2). Our observations demonstrate that exogenous VEGF stimulates GBSC proliferation in a dose-dependent manner via VEGF Receptor 2 (VEGFR2); while VEGF Receptor 1 (VEGFR1) has a negative feedback effect on VEGFR2 when cells were exposed to higher concentration of VEGF. These results suggest that suppressing VEGFR2-dependent GBSC proliferation is a potentially therapeutic strategy in GBM.
Collapse
|