1
|
Taranov A, Bedolla A, Iwasawa E, Brown FN, Baumgartner S, Fugate EM, Levoy J, Crone SA, Goto J, Luo Y. The choroid plexus maintains adult brain ventricles and subventricular zone neuroblast pool, which facilitates poststroke neurogenesis. Proc Natl Acad Sci U S A 2024; 121:e2400213121. [PMID: 38954546 PMCID: PMC11252789 DOI: 10.1073/pnas.2400213121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
The brain's neuroreparative capacity after injuries such as ischemic stroke is partly contained in the brain's neurogenic niches, primarily the subventricular zone (SVZ), which lies in close contact with the cerebrospinal fluid (CSF) produced by the choroid plexus (ChP). Despite the wide range of their proposed functions, the ChP/CSF remain among the most understudied compartments of the central nervous system (CNS). Here, we report a mouse genetic tool (the ROSA26iDTR mouse line) for noninvasive, specific, and temporally controllable ablation of CSF-producing ChP epithelial cells to assess the roles of the ChP and CSF in brain homeostasis and injury. Using this model, we demonstrate that ChP ablation causes rapid and permanent CSF volume loss in both aged and young adult brains, accompanied by disruption of ependymal cilia bundles. Surprisingly, ChP ablation did not result in overt neurological deficits at 1 mo postablation. However, we observed a pronounced decrease in the pool of SVZ neuroblasts (NBs) following ChP ablation, which occurs due to their enhanced migration into the olfactory bulb. In the middle cerebral artery occlusion model of ischemic stroke, NB migration into the lesion site was also reduced in the CSF-depleted mice. Thus, our study establishes an important role of ChP/CSF in regulating the regenerative capacity of the adult brain under normal conditions and after ischemic stroke.
Collapse
Affiliation(s)
- Aleksandr Taranov
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH45229
| | - Alicia Bedolla
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH45229
| | - Eri Iwasawa
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Farrah N. Brown
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Sarah Baumgartner
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Elizabeth M. Fugate
- Imaging Research Center, Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH45229
| | - Joel Levoy
- Imaging Research Center, Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH45229
| | - Steven A. Crone
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Neurosurgery, College of Medicine, University of Cincinnati, Cincinnati, OH45267
| | - June Goto
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Neurosurgery, College of Medicine, University of Cincinnati, Cincinnati, OH45267
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH45229
| |
Collapse
|
2
|
Bedolla A, Wegman E, Weed M, Stevens MK, Ware K, Paranjpe A, Alkhimovitch A, Ifergan I, Taranov A, Peter JD, Gonzalez RMS, Robinson JE, McClain L, Roskin KM, Greig NH, Luo Y. Adult microglial TGFβ1 is required for microglia homeostasis via an autocrine mechanism to maintain cognitive function in mice. Nat Commun 2024; 15:5306. [PMID: 38906887 PMCID: PMC11192737 DOI: 10.1038/s41467-024-49596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
While TGF-β signaling is essential for microglial function, the cellular source of TGF-β1 ligand and its spatial regulation remains unclear in the adult CNS. Our data supports that microglia but not astrocytes or neurons are the primary producers of TGF-β1 ligands needed for microglial homeostasis. Microglia-Tgfb1 KO leads to the activation of microglia featuring a dyshomeostatic transcriptome that resembles disease-associated, injury-associated, and aged microglia, suggesting microglial self-produced TGF-β1 ligands are important in the adult CNS. Astrocytes in MG-Tgfb1 inducible (i)KO mice show a transcriptome profile that is closely aligned with an LPS-associated astrocyte profile. Additionally, using sparse mosaic single-cell microglia KO of TGF-β1 ligand we established an autocrine mechanism for signaling. Here we show that MG-Tgfb1 iKO mice present cognitive deficits, supporting that precise spatial regulation of TGF-β1 ligand derived from microglia is required for the maintenance of brain homeostasis and normal cognitive function in the adult brain.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Max Weed
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | | | - Kierra Ware
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Information Services for Research, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Anastasia Alkhimovitch
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Igal Ifergan
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Aleksandr Taranov
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Joshua D Peter
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Rosa Maria Salazar Gonzalez
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
| | - J Elliott Robinson
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
| | - Lucas McClain
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Krishna M Roskin
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA.
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA.
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
3
|
Michinaga S, Hishinuma S, Koyama Y. Roles of astrocytic sonic hedgehog production and its signal for regulation of the blood-brain barrier permeability. VITAMINS AND HORMONES 2024; 126:97-111. [PMID: 39029978 DOI: 10.1016/bs.vh.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Sonic hedgehog (Shh) is a secreted glycopeptide belonging to the hedgehog family that is essential for morphogenesis during embryonic development. The Shh signal is mediated by two membrane proteins, Patched-1 (Ptch-1) and Smoothened (Smo), following the activation of transcription factors such as Gli. Shh decreases the permeability of the blood-brain barrier (BBB) and plays a key role in its function. In the damaged brain, BBB function is remarkably disrupted. The BBB disruption causes brain edema and neuroinflammation resulting from the extravasation of serum components and the infiltration of inflammatory cells into the cerebral parenchyma. Multiple studies have suggested that astrocyte is a source of Shh and that astrocytic Shh production is increased in the damaged brain. In various experimental animal models of acute brain injury, Shh or Shh signal activators alleviate BBB disruption by increasing tight junction proteins in endothelial cells. Furthermore, activation of astrocytic Shh signaling reduces reactive astrogliosis, neuroinflammation, and increases the production of vascular protective factors, which alleviates BBB disruption in the damaged brain. These findings suggest that astrocytic Shh and Shh signaling protect BBB function in the damaged brain and that target drugs for Shh signaling are expected to be novel therapeutic drugs for acute brain injuries.
Collapse
Affiliation(s)
- Shotaro Michinaga
- Department of Pharmacodynamics, Meiji Pharmaceutical University, Tokyo, Japan
| | - Shigeru Hishinuma
- Department of Pharmacodynamics, Meiji Pharmaceutical University, Tokyo, Japan
| | - Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan.
| |
Collapse
|
4
|
Li Y, Xue W, Li S, Cui L, Gao Y, Li L, Chen R, Zhang X, Xu R, Jiang W, Zhang X, Wang L. Salidroside promotes angiogenesis after cerebral ischemia in mice through Shh signaling pathway. Biomed Pharmacother 2024; 174:116625. [PMID: 38643543 DOI: 10.1016/j.biopha.2024.116625] [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: 12/19/2023] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024] Open
Abstract
AIMS The purpose of this study was to explore the impacts of salidroside on vascular regeneration, vascular structural changes and long-term neurological recuperation following cerebral ischemia and its possible mechanism. MAIN METHODS From Day 1 to Day 28, young male mice with middle cerebral artery blockage received daily doses of salidroside and measured neurological deficits. On the 7th day after stroke, the volume of cerebral infarction was determined using TTC and HE staining. Microvascular density, astrocyte coverage, angiogenesis and the expression of the Shh signaling pathway were detected by IF, qRTPCR and WB at 7, 14 and 28 days after stroke. Changes in blood flow, blood vessel density and diameter from stroke to 28 days were measured by the LSCI and TPMI. KEY FINDINGS Compared with the dMACO group, the salidroside treatment group significantly promoted the recovery of neurological function. Salidroside was found to enhance cerebral blood flow perfusion and reduce the infarct on the 7th day after stroke. From the 7th to the 28th day after stroke, salidroside treatment boosted the expression of CD31, CD31+/BrdU+, and GFAP in the cortex around the infarction site. On the 14th day after stroke, salidroside significantly enhanced the width and density of blood vessels. Salidroside increased the expression of histones and genes in the Shh signaling pathway during treatment, and this effect was weakened by the Shh inhibitor Cyclopamine. SIGNIFICANCE Salidroside can restore nerve function, improve cerebral blood flow, reduce cerebral infarction volume, increase microvessel density and promote angiogenesis via the Shh signaling pathway.
Collapse
Affiliation(s)
- Ying Li
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Weihong Xue
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Songyi Li
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Lili Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yuxiao Gao
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Linlin Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Rong Chen
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiao Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Renhao Xu
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Wei Jiang
- Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China.
| | - Lina Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Collaborative Innovation Center for Cardio, Cerebrovascular Disease, Shijiazhuang, Hebei 050000, People's Republic of China; Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, Hebei 050000, People's Republic of China.
| |
Collapse
|
5
|
Taranov A, Bedolla A, Iwasawa E, Brown FN, Baumgartner S, Fugate EM, Levoy J, Crone SA, Goto J, Luo Y. The choroid plexus maintains ventricle volume and adult subventricular zone neuroblast pool, which facilitates post-stroke neurogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.575277. [PMID: 38328050 PMCID: PMC10849542 DOI: 10.1101/2024.01.22.575277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The brain's neuroreparative capacity after injuries such as ischemic stroke is contained in the brain's neurogenic niches, primarily the subventricular zone (SVZ), which lies in close contact with the cerebrospinal fluid (CSF) produced by the choroid plexus (ChP). Despite the wide range of their proposed functions, the ChP/CSF remain among the most understudied compartments of the central nervous system (CNS). Here we report a mouse genetic tool (the ROSA26iDTR mouse line) for non-invasive, specific, and temporally controllable ablation of CSF-producing ChP epithelial cells to assess the roles of the ChP and CSF in brain homeostasis and injury. Using this model, we demonstrate that ChP ablation causes rapid and permanent CSF volume loss accompanied by disruption of ependymal cilia bundles. Surprisingly, ChP ablation did not result in overt neurological deficits at one-month post-ablation. However, we observed a pronounced decrease in the pool of SVZ neuroblasts following ChP ablation, which occurs due to their enhanced migration into the olfactory bulb. In the MCAo model of ischemic stroke, neuroblast migration into the lesion site was also reduced in the CSF-depleted mice. Thus, our study establishes an important and novel role of ChP/CSF in regulating the regenerative capacity of the adult brain under normal conditions and after ischemic stroke.
Collapse
Affiliation(s)
- Aleksandr Taranov
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Alicia Bedolla
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Eri Iwasawa
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Farrah N. Brown
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Sarah Baumgartner
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Elizabeth M. Fugate
- Imaging Research Center, Cincinnati Children’s Hospital Medical Center, Department of Radiology, University of Cincinnati, Cincinnati, USA
| | - Joel Levoy
- Imaging Research Center, Cincinnati Children’s Hospital Medical Center, Department of Radiology, University of Cincinnati, Cincinnati, USA
| | - Steven A. Crone
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Neurosurgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - June Goto
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Neurosurgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
6
|
Bedolla AM, McKinsey GL, Ware K, Santander N, Arnold TD, Luo Y. A comparative evaluation of the strengths and potential caveats of the microglial inducible CreER mouse models. Cell Rep 2024; 43:113660. [PMID: 38217856 PMCID: PMC10874587 DOI: 10.1016/j.celrep.2023.113660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/02/2023] [Accepted: 12/20/2023] [Indexed: 01/15/2024] Open
Abstract
The recent proliferation of new Cre and CreER recombinase lines provides researchers with a diverse toolkit to study microglial gene function. To determine how best to apply these lines in studies of microglial gene function, a thorough and detailed comparison of their properties is needed. Here, we examined four different microglial CreER lines (Cx3cr1YFP-CreER(Litt), Cx3cr1CreER(Jung), P2ry12CreER, and Tmem119CreER), focusing on (1) recombination specificity, (2) leakiness (the degree of tamoxifen-independent recombination in microglia and other cells), (3) the efficiency of tamoxifen-induced recombination, (4) extraneural recombination (the degree of recombination in cells outside of the CNS, particularly myelo/monocyte lineages), and (5) off-target effects in the context of neonatal brain development. We identify important caveats and strengths for these lines, which will provide broad significance for researchers interested in performing conditional gene deletion in microglia. We also provide data emphasizing the potential of these lines for injury models that result in the recruitment of splenic immune cells.
Collapse
Affiliation(s)
- Alicia M Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Gabriel L McKinsey
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kierra Ware
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Nicolas Santander
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center.
| |
Collapse
|
7
|
Hussain G, Akram R, Anwar H, Sajid F, Iman T, Han HS, Raza C, De Aguilar JLG. Adult neurogenesis: a real hope or a delusion? Neural Regen Res 2024; 19:6-15. [PMID: 37488837 PMCID: PMC10479850 DOI: 10.4103/1673-5374.375317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/27/2023] [Accepted: 04/10/2023] [Indexed: 07/26/2023] Open
Abstract
Adult neurogenesis, the process of creating new neurons, involves the coordinated division, migration, and differentiation of neural stem cells. This process is restricted to neurogenic niches located in two distinct areas of the brain: the subgranular zone of the dentate gyrus of the hippocampus and the subventricular zone of the lateral ventricle, where new neurons are generated and then migrate to the olfactory bulb. Neurogenesis has been thought to occur only during the embryonic and early postnatal stages and to decline with age due to a continuous depletion of neural stem cells. Interestingly, recent years have seen tremendous progress in our understanding of adult brain neurogenesis, bridging the knowledge gap between embryonic and adult neurogenesis. Here, we discuss the current status of adult brain neurogenesis in light of what we know about neural stem cells. In this notion, we talk about the importance of intracellular signaling molecules in mobilizing endogenous neural stem cell proliferation. Based on the current understanding, we can declare that these molecules play a role in targeting neurogenesis in the mature brain. However, to achieve this goal, we need to avoid the undesired proliferation of neural stem cells by controlling the necessary checkpoints, which can lead to tumorigenesis and prove to be a curse instead of a blessing or hope.
Collapse
Affiliation(s)
- Ghulam Hussain
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Rabia Akram
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Haseeb Anwar
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Faiqa Sajid
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Tehreem Iman
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Punjab, Pakistan
| | - Hyung Soo Han
- Department of Physiology, School of Medicine, Clinical Omics Institute, Kyungpook National University, Daegu, Korea
| | - Chand Raza
- Department of Zoology, Faculty of Chemistry and Life Sciences, Government College University, Lahore, Pakistan
| | - Jose-Luis Gonzalez De Aguilar
- INSERM, U1118, Mécanismes Centraux et Péripheriques de la Neurodégénérescence, Strasbourg, France, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
8
|
Dai MJ, Gui XX, Jia SM, Lv ST, Dou H, Cui W. Dl-3-n-butylphthalide promotes angiogenesis in ischemic stroke mice through upregulating autocrine and paracrine sonic hedgehog. Acta Pharmacol Sin 2023; 44:2404-2417. [PMID: 37580491 PMCID: PMC10692133 DOI: 10.1038/s41401-023-01137-z] [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: 02/17/2023] [Accepted: 07/09/2023] [Indexed: 08/16/2023] Open
Abstract
Dl-3-n-butylphthalide (NBP) is a small-molecule drug used in the treatment of ischemic stroke in China, which is proven to ameliorate the symptoms of ischemic stroke and improve the prognosis of patients. Previous studies have shown that NBP accelerates recovery after stroke by promoting angiogenesis. In this study, we investigated the mechanisms underlying the angiogenesis-promoting effects of NBP in ischemic stroke models in vitro and in vivo. OGD/R model was established in human umbilical vein endothelial cells (HUVECs) and human brain microvascular endothelial cells (HBMECs), while the tMCAO model was established in mice. The cells were pretreated with NBP (10, 50, 100 µM); the mice were administered NBP (4, 8 mg/kg, i.v.) twice after tMCAO. We showed that NBP treatment significantly stimulated angiogenesis by inducing massive production of angiogenic growth factors VEGFA and CD31 in both in vitro and in vivo models of ischemic stroke. NBP also increased the tubule formation rate and migration capability of HUVECs in vitro. By conducting the weighted gene co-expression network analysis, we found that these effects were achieved by upregulating the expression of a hedgehog signaling pathway. We demonstrated that NBP treatment not only changed the levels of regulators of the hedgehog signaling pathway but also activated the transcription factor Gli1. The pro-angiogenesis effect of NBP was abolished when the hedgehog signaling pathway was inhibited by GDC-0449 in HUVECs, by Sonic Hedgehog(Shh) knockdown in HUVECs, or by intracerebroventricular injection of AAV-shRNA(shh)-CMV in tMCAO mice. Furthermore, we found that HUVECs produced a pro-angiogenic response not only to autocrine Shh, but also to paracrine Shh secreted by astrocytes. Together, we demonstrate that NBP promotes angiogenesis via upregulating the hedgehog signaling pathway. Our results provide an experimental basis for the clinical use of NBP.
Collapse
Affiliation(s)
- Mei-Jie Dai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xing-Xing Gui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Shu-Miao Jia
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Shu-Ting Lv
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hao Dou
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wei Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
9
|
Li MR, Luo XJ, Peng J. Role of sonic hedgehog signaling pathway in the regulation of ion channels: focus on its association with cardio-cerebrovascular diseases. J Physiol Biochem 2023; 79:719-730. [PMID: 37676576 DOI: 10.1007/s13105-023-00982-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Sonic hedgehog (SHH) signaling is vital for cell differentiation and proliferation during embryonic development, yet its role in cardiac, cerebral, and vascular pathophysiology is under debate. Recent studies have demonstrated that several compounds of SHH signaling regulate ion channels, which in turn affect the behavior of target cells. Some of these ion channels are involved in the cardio-cerebrovascular system. Here, we first reviewed the SHH signaling cascades, then its interaction with ion channels, and their impact on cardio-cerebrovascular diseases. Considering the complex cross talk of SHH signaling with other pathways that also affect ion channels and their potential impact on the cardio-cerebrovascular system, we highlight the necessity of thoroughly studying the effect of SHH signaling on ion homeostasis, which could serve as a novel mechanism for cardio-cerebrovascular diseases. Activation of SHH signaling influence ion channels activity, which in turn influence ion homeostasis, membrane potential, and electrophysiology, could serve as a novel strategy for cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
10
|
Zhao X, Zhu J, Chen S, Liu R, Long T. Neural Stem Cell-Derived Exosomes Improve Neurological Function in Rats with Cerebral Ischemia-Reperfusion Injury by Regulating Microglia-Mediated Inflammatory Response. J Inflamm Res 2023; 16:3079-3092. [PMID: 37520663 PMCID: PMC10378531 DOI: 10.2147/jir.s414121] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/08/2023] [Indexed: 08/01/2023] Open
Abstract
Purpose To investigate the effect of neural stem cell-derived exosomes (NSC-Exos) on neural function after rat cerebral ischemia-reperfusion injury by regulating microglia-mediated inflammatory response. Methods SD rats were randomly divided into Sham group, IRI group, PBS group and NSC-Exos group. Each group was divided into 1d, 3d, 7d and 14d subgroups. In the Sham group, only cervical vessels were isolated without blockage. MCAO model was constructed in the other three groups by blocking middle cerebral artery with thread embolism. PBS group and NSC-Exos group were, respectively, injected into the lateral ventricle of PBS and Exos. Neurobehavioral deficit scores were performed for each subgroup at relative time points, then brains were taken for TTC staining, parietal cortex histopathology and microglia-mediated inflammatory response-related factors were detected. Results Compared with Sham group, neurological defect score and infarction volume in both the IRI and PBS groups were significantly increased. The exploration target quadrant time and escape latency time of maze test were increased. The number of CD86+/Iba1+ double-positive cells increased, while CD206+/Iba1+ double-positive cells decreased. The expressions of IL-6 and CD86 in parietal cortex were increased, while the expressions of Arg1 and CD206 were decreased. Compared with the IRI group and PBS group, neurological defect score and infarction volume in NSC-Exos group were decreased. The exploration target quadrant time and escape latency time of water maze test were decreased. The number of CD206+/Iba1+ double-positive cells increased, while CD86+/Iba1+ double-positive cells decreased. The expressions of Arg1 and CD206 in parietal cortex were increased, while the expressions of IL-6 and CD86 were decreased. Conclusion NSC-Exos can promote the polarization of microglia, that is, inhibit the polarization of M1 and promote polarization of M2, reduce microglia-mediated neuroinflammation, suggesting that NSC-Exos may be a strategy for the treatment of microglia-mediated neuroinflammation after ischemic brain injury.
Collapse
Affiliation(s)
- Xue Zhao
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Junde Zhu
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Shan Chen
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Ruojing Liu
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Tingting Long
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| |
Collapse
|
11
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
12
|
Prajapati A, Mehan S, Khan Z. The role of Smo-Shh/Gli signaling activation in the prevention of neurological and ageing disorders. Biogerontology 2023:10.1007/s10522-023-10034-1. [PMID: 37097427 DOI: 10.1007/s10522-023-10034-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/05/2023] [Indexed: 04/26/2023]
Abstract
Sonic hedgehog (Shh) signaling is an essential central nervous system (CNS) pathway involved during embryonic development and later life stages. Further, it regulates cell division, cellular differentiation, and neuronal integrity. During CNS development, Smo-Shh signaling is significant in the proliferation of neuronal cells such as oligodendrocytes and glial cells. The initiation of the downstream signalling cascade through the 7-transmembrane protein Smoothened (Smo) promotes neuroprotection and restoration during neurological disorders. The dysregulation of Smo-Shh is linked to the proteolytic cleavage of GLI (glioma-associated homolog) into GLI3 (repressor), which suppresses target gene expression, leading to the disruption of cell growth processes. Smo-Shh aberrant signalling is responsible for several neurological complications contributing to physiological alterations like increased oxidative stress, neuronal excitotoxicity, neuroinflammation, and apoptosis. Moreover, activating Shh receptors in the brain promotes axonal elongation and increases neurotransmitters released from presynaptic terminals, thereby exerting neurogenesis, anti-oxidation, anti-inflammatory, and autophagy responses. Smo-Shh activators have been shown in preclinical and clinical studies to help prevent various neurodegenerative and neuropsychiatric disorders. Redox signalling has been found to play a critical role in regulating the activity of the Smo-Shh pathway and influencing downstream signalling events. In the current study ROS, a signalling molecule, was also essential in modulating the SMO-SHH gli signaling pathway in neurodegeneration. As a result of this investigation, dysregulation of the pathway contributes to the pathogenesis of various neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD).Thus, Smo-Shh signalling activators could be a potential therapeutic intervention to treat neurocomplications of brain disorders.
Collapse
Affiliation(s)
- Aradhana Prajapati
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
13
|
Bedolla A, Mckinsey G, Ware K, Santander N, Arnold T, Luo Y. Finding the right tool: a comprehensive evaluation of microglial inducible cre mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.536878. [PMID: 37131606 PMCID: PMC10153116 DOI: 10.1101/2023.04.17.536878] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The recent proliferation of new Cre and CreER recombinase lines provides researchers with a diverse toolkit to study microglial gene function. To determine how best to apply these lines in studies of microglial gene function, a thorough and detailed comparison of their properties is needed. Here, we examined four different microglial CreER lines (Cx3cr1CreER(Litt), Cx3cr1CreER(Jung), P2ry12CreER, Tmem119CreER), focusing on (1) recombination specificity; (2) leakiness - degree of non-tamoxifen recombination in microglia and other cells; (3) efficiency of tamoxifen-induced recombination; (4) extra-neural recombination -the degree of recombination in cells outside the CNS, particularly myelo/monocyte lineages (5) off-target effects in the context of neonatal brain development. We identify important caveats and strengths for these lines which will provide broad significance for researchers interested in performing conditional gene deletion in microglia. We also provide data emphasizing the potential of these lines for injury models that result in the recruitment of splenic immune cells.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Gabriel Mckinsey
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kierra Ware
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Nicolas Santander
- Instituto de Ciencias de la Salud, Universidad de O´Higgins, Rancagua, Chile
| | - Thomas Arnold
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
14
|
Dlamini Z, Khanyile R, Molefi T, Damane BP, Bates DO, Hull R. Genomic Interplay between Neoneurogenesis and Neoangiogenesis in Carcinogenesis: Therapeutic Interventions. Cancers (Basel) 2023; 15:cancers15061805. [PMID: 36980690 PMCID: PMC10046518 DOI: 10.3390/cancers15061805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Angiogenesis, the generation of new blood vessels, is one of the hallmarks of cancer. The growing tumor requires nutrients and oxygen. Recent evidence has shown that tumors release signals to attract new nerve fibers and stimulate the growth of new nerve fibers. Neurogenesis, neural extension, and axonogenesis assist in the migration of cancer cells. Cancer cells can use both blood vessels and nerve fibers as routes for cells to move along. In this way, neurogenesis and angiogenesis both contribute to cancer metastasis. As a result, tumor-induced neurogenesis joins angiogenesis and immunosuppression as aberrant processes that are exacerbated within the tumor microenvironment. The relationship between these processes contributes to cancer development and progression. The interplay between these systems is brought about by cytokines, neurotransmitters, and neuromodulators, which activate signaling pathways that are common to angiogenesis and the nervous tissue. These include the AKT signaling pathways, the MAPK pathway, and the Ras signaling pathway. These processes also both require the remodeling of tissues. The interplay of these processes in cancer provides the opportunity to develop novel therapies that can be used to target these processes.
Collapse
Affiliation(s)
- Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0028, South Africa
- Correspondence: (Z.D.); (R.H.)
| | - Richard Khanyile
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0028, South Africa
- Department of Medical Oncology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa
| | - Thulo Molefi
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0028, South Africa
- Department of Medical Oncology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa
| | - Botle Precious Damane
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa
| | - David Owen Bates
- Centre for Cancer Sciences, Division of Cancer and Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0028, South Africa
- Correspondence: (Z.D.); (R.H.)
| |
Collapse
|
15
|
Prospective Randomized Control Trial to Compare the Role of Injection Cerebrolysin for 10 Days Duration Against Placebo in Operated Cases of Degenerative Cervical Myelopathy. Spine (Phila Pa 1976) 2023; 48:295-300. [PMID: 36730671 DOI: 10.1097/brs.0000000000004542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/08/2022] [Indexed: 02/04/2023]
Abstract
STUDY DESIGN Prospective randomized control trial. OBJECTIVE To analyze outcomes following the injection of cerebrolysin in surgically treated patients with degenerative cervical myelopathy (DCM). SUMMARY OF BACKGROUND DATA Previous research has concluded that superior functional outcomes are achieved with the use of cerebrolysin in surgically treated patients of DCM for 21 days. Our study has been conducted to analyze the use of this drug for a shorter duration (10 days) and compare its clinical efficacy. METHODS Ninety operated cases of mild to severe DCM were randomized into two groups. Sixty patients received the injection Cerebrolysin for 10 days postoperatively. The remaining 30 patients received a placebo. Functional outcomes were measured using modified Japanese Orthopaedic Association (mJOA) scores and visual analogue scale (VAS). The American Spinal Injury Association (ASIA) scale was used to document neurological recovery. Hand function was assessed by measuring the grip strength and the upper limb function score the upper extremity motor mJOA plus upper extremity sensory mJOA score. Assessments were performed and preoperatively and postoperatively and at one-month, three-month, six-month, and one-year following surgery. RESULTS Preoperative mJOA and VAS scores were comparable in both groups ( P >0.05). Both groups experienced an improvement in mJOA and VAS scores at all time-points during follow-up as compared with preoperative scores. However, the cerebrolysin group demonstrated significantly greater mJOA scores (16.37±1) when compared with the placebo (15.2±1.8) at one-year follow-up ( P <0.0001). Neurological improvement with cerebrolysin therapy was also superior ( P =0.04). No significant adverse reactions were documented. CONCLUSION Injection cerebrolysin, when administered for 10 days postoperatively, can result in significantly greater neurological improvement and hand function in patients with DCM who also receive surgery.
Collapse
|
16
|
Phillips CM, Stamatovic SM, Keep RF, Andjelkovic AV. Epigenetics and stroke: role of DNA methylation and effect of aging on blood-brain barrier recovery. Fluids Barriers CNS 2023; 20:14. [PMID: 36855111 PMCID: PMC9972738 DOI: 10.1186/s12987-023-00414-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/10/2023] [Indexed: 03/02/2023] Open
Abstract
Incomplete recovery of blood-brain barrier (BBB) function contributes to stroke outcomes. How the BBB recovers after stroke remains largely unknown. Emerging evidence suggests that epigenetic factors play a significant role in regulating post-stroke BBB recovery. This study aimed to evaluate the epigenetic and transcriptional profile of cerebral microvessels after thromboembolic (TE) stroke to define potential causes of limited BBB recovery. RNA-sequencing and reduced representation bisulfite sequencing (RRBS) analyses were performed using microvessels isolated from young (6 months) and old (18 months) mice seven days poststroke compared to age-matched sham controls. DNA methylation profiling of poststroke brain microvessels revealed 11,287 differentially methylated regions (DMR) in old and 9818 DMR in young mice, corresponding to annotated genes. These DMR were enriched in genes encoding cell structural proteins (e.g., cell junction, and cell polarity, actin cytoskeleton, extracellular matrix), transporters and channels (e.g., potassium transmembrane transporter, organic anion and inorganic cation transporters, calcium ion transport), and proteins involved in endothelial cell processes (e.g., angiogenesis/vasculogenesis, cell signaling and transcription regulation). Integrated analysis of methylation and RNA sequencing identified changes in cell junctions (occludin), actin remodeling (ezrin) as well as signaling pathways like Rho GTPase (RhoA and Cdc42ep4). Aging as a hub of aberrant methylation affected BBB recovery processes by profound alterations (hypermethylation and repression) in structural protein expression (e.g., claudin-5) as well as activation of a set of genes involved in endothelial to mesenchymal transformation (e.g., Sox9, Snai1), repression of angiogenesis and epigenetic regulation. These findings revealed that DNA methylation plays an important role in regulating BBB repair after stroke, through regulating processes associated with BBB restoration and prevalently with processes enhancing BBB injury.
Collapse
Affiliation(s)
- Chelsea M Phillips
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Svetlana M Stamatovic
- Department of Pathology, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Richard F Keep
- Department of Neurosurgery, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Pathology, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA. .,Department of Neurosurgery, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.
| |
Collapse
|
17
|
Cheng B, Sharma DR, Kumar A, Sheth H, Agyemang A, Aschner M, Zhang X, Ballabh P. Shh activation restores interneurons and cognitive function in newborns with intraventricular haemorrhage. Brain 2023; 146:629-644. [PMID: 35867870 PMCID: PMC10169407 DOI: 10.1093/brain/awac271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/27/2022] [Accepted: 07/05/2022] [Indexed: 11/14/2022] Open
Abstract
Premature infants with germinal matrix haemorrhage-intraventricular haemorrhage (GMH-IVH) suffer from neurobehavioural deficits as they enter childhood and adolescence. Yet the underlying mechanisms remain unclear. Impaired development and function of interneurons contribute to neuropsychiatric disorders. Therefore, we hypothesized that the occurrence of IVH would reduce interneuron neurogenesis in the medial ganglionic eminence and diminish the population of parvalbumin+ and somatostatin+ cortical interneurons. Because Sonic Hedgehog promotes the production of cortical interneurons, we also postulated that the activation of Sonic Hedgehog signalling might restore neurogenesis, cortical interneuron population, and neurobehavioural function in premature newborns with IVH. These hypotheses were tested in a preterm rabbit model of IVH and autopsy samples from human preterm infants. We compared premature newborns with and without IVH for intraneuronal progenitors, cortical interneurons, transcription factors regulating neurogenesis, single-cell transcriptome of medial ganglionic eminence and neurobehavioural functions. We treated premature rabbit kits with adenovirus expressing Sonic Hedgehog (Ad-Shh) or green fluorescence protein gene to determine the effect of Sonic Hedgehog activation on the interneuron production, cortical interneuron population and neurobehaviour. We discovered that IVH reduced the number of Nkx2.1+ and Dlx2+ progenitors in the medial ganglionic eminence of both humans and rabbits by attenuating their proliferation and inducing apoptosis. Moreover, IVH decreased the population of parvalbumin+ and somatostatin+ neurons in the frontal cortex of both preterm infants and kits relative to controls. Sonic Hedgehog expression and the downstream transcription factors, including Nkx2.1, Mash1, Lhx6 and Sox6, were also reduced in kits with IVH. Consistent with these findings, single-cell transcriptomic analyses of medial ganglionic eminence identified a distinct subpopulation of cells exhibiting perturbation in genes regulating neurogenesis, ciliogenesis, mitochondrial function and MAPK signalling in rabbits with IVH. More importantly, restoration of Sonic Hedgehog level by Ad-Shh treatment ameliorated neurogenesis, cortical interneuron population and neurobehavioural function in kits with IVH. Additionally, Sonic Hedgehog activation alleviated IVH-induced inflammation and several transcriptomic changes in the medial ganglionic eminence. Taken together, IVH reduced intraneuronal production and cortical interneuron population by downregulating Sonic Hedgehog signalling in both preterm rabbits and humans. Notably, activation of Sonic Hedgehog signalling restored interneuron neurogenesis, cortical interneurons and cognitive function in rabbit kits with IVH. These findings highlight disruption in cortical interneurons in IVH and identify a novel therapeutic strategy to restore cortical interneurons and cognitive function in infants with IVH. These studies can accelerate the development of new therapies to enhance the neurodevelopmental outcome of survivors with IVH.
Collapse
Affiliation(s)
- Bokun Cheng
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deep R Sharma
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ajeet Kumar
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hardik Sheth
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alex Agyemang
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Aschner
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xusheng Zhang
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
18
|
Phillips C, Stamatovic S, Keep R, Andjelkovic A. Epigenetics and stroke: role of DNA methylation and effect of aging on blood-brain barrier recovery. RESEARCH SQUARE 2023:rs.3.rs-2444060. [PMID: 36711725 PMCID: PMC9882686 DOI: 10.21203/rs.3.rs-2444060/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Incomplete recovery of blood-brain barrier (BBB) function contributes to stroke outcomes. How the BBB recovers after stroke remains largely unknown. Emerging evidence suggests that epigenetic factors play a significant role in regulating post-stroke BBB recovery. This study aimed to evaluate the epigenetic and transcriptional profile of cerebral microvessels after thromboembolic (TE) stroke to define potential causes of limited BBB recovery. RNA-sequencing and reduced representation bisulfite sequencing (RRBS) analyses were performed using microvessels isolated from young (6 months) and old (18 months) mice seven days poststroke compared to age-matched sham controls. DNA methylation profiling of poststroke brain microvessels revealed 11287 differentially methylated regions (DMR) in old and 9818 DMR in young mice, corresponding to annotated genes. These DMR were enriched in genes encoding cell structural proteins (e.g., cell junction, and cell polarity, actin cytoskeleton, extracellular matrix), transporters and channels (e.g., potassium transmembrane transporter, organic anion and inorganic cation transporters, calcium ion transport), and proteins involved in endothelial cell processes (e.g., angiogenesis/vasculogenesis, cell signaling and transcription regulation). Integrated analysis of methylation and RNA sequencing identified changes in cell junctions (occludin), actin remodeling (ezrin) as well as signaling pathways like Rho GTPase (RhoA and Cdc42ep4). Aging as a hub of aberrant methylation affected BBB recovery processes by profound alterations (hypermethylation and repression) in structural protein expression (e.g., claudin-5) as well as activation of a set of genes involved in endothelial to mesenchymal transformation (e.g., Sox17 , Snail1 ), repression of angiogenesis and epigenetic regulation. These findings revealed that DNA methylation plays an important role in regulating BBB repair after stroke, through regulating processes associated with BBB restoration and prevalently with processes enhancing BBB injury.
Collapse
|
19
|
Yu CC, Liu LB, Chen SY, Wang XF, Wang L, Du YJ. Ancient Chinese Herbal Recipe Huanglian Jie Du Decoction for Ischemic Stroke: An Overview of Current Evidence. Aging Dis 2022; 13:1733-1744. [PMID: 36465168 PMCID: PMC9662271 DOI: 10.14336/ad.2022.0311] [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: 02/13/2022] [Accepted: 03/11/2022] [Indexed: 09/30/2023] Open
Abstract
Ischemic stroke is a major cause of mortality and neurological morbidity worldwide. The underlying pathophysiology of ischemic stroke is highly complicated and correlates with various pathological processes, including neuroinflammation, oxidative stress injury, altered cell apoptosis and autophagy, excitotoxicity, and acidosis. The current treatment for ischemic stroke is limited to thrombolytic therapy such as recombinant tissue plasminogen activator. However, tissue plasminogen activator is limited by a very narrow therapeutic time window (<4.5 hours), selective efficacy, and hemorrhagic complication. Hence, the development of novel therapies to prevent ischemic damage to the brain is urgent. Chinese herbal medicine has a long history in treating stroke and its sequela. In the past decades, extensive studies have focused on the neuroprotective effects of Huanglian Jie Du decoction (HLJDD), an ancient and classical Chinese herbal formula that can treat a wide spectrum of disorders including ischemic stroke. In this review, the current evidence of HLJDD and its bioactive components for ischemic stroke is comprehensively reviewed, and their potential application directions in ischemic stroke management are discussed.
Collapse
Affiliation(s)
- Chao-Chao Yu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
| | - Le-Bin Liu
- Department of Rehabilitation Medicine, Hubei Rongjun Hospital, Wuhan, Hubei, China.
| | - Shi-Yuan Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Xiao-Fei Wang
- Department of Rehabilitation Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Li Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
| |
Collapse
|
20
|
Gupta R, Mehan S, Chhabra S, Giri A, Sherawat K. Role of Sonic Hedgehog Signaling Activation in the Prevention of Neurological Abnormalities Associated with Obsessive-Compulsive Disorder. Neurotox Res 2022; 40:1718-1738. [PMID: 36272053 DOI: 10.1007/s12640-022-00586-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 12/31/2022]
Abstract
The smoothened sonic hedgehog (Smo-Shh) pathway is one mechanism that influences neurogenesis, including brain cell differentiation and development during childhood. Shh signaling dysregulation leads to decreased target gene transcription, which contributes to increased neuronal excitation, apoptosis, and neurodegeneration, eventually leading to neurological deficits. Neuropsychiatric disorders such as OCD and related neurological dysfunctions are characterized by neurotransmitter imbalance, neuroinflammation, oxidative stress, and impaired neurogenesis, disturbing the cortico-striato-thalamo-cortical (CSTC) link neuronal network. Despite the availability of several treatments, such as selective serotonin reuptake inhibitors, some individuals may not benefit much from them. Several trials on the use of antipsychotics in the treatment of OCD have also produced inadequate findings. This evidence-based review focuses on a potential pharmacological approach to alleviating OCD and associated neuronal deficits by preventing neurochemical alterations, in which sonic hedgehog activators are neuroprotective, lowering neuronal damage while increasing neuronal maintenance and survival. As a result, stimulating SMO-Shh via its potential activators may have neuroprotective effects on neurological impairment associated with OCD. This review investigates the link between SMO-Shh signaling and the neurochemical abnormalities associated with the progression of OCD and associated neurological dysfunctions. Role of Smo-Shh signaling in serotonergic neurogenesis and in maintaining their neuronal identity. The Shh ligand activates two main transcriptional factors known as Foxa2 and Nkx2.2, which again activates another transcriptional factor, GATA (GATA2 and GATA3), in post mitotic precursor cells of serotonergic neurons-following increased expression of Pet-1 and Lmx1b after GATA regulates the expression of many serotonergic enzymes such as TPH2, SERT, VMAT, slc6a4, Htr1a, Htr1b (Serotonin receptor enzymes), and MAO that regulate and control the release of serotonin and maintain their neuronal identity after their maturation. Abbreviation: Foxa2: Forkhead box; GATA: Globin transcription factor; Lmx1b: LIM homeobox transcription factor 1 beta; TPH2: Tryptophan hydroxylase 2; Htr1a: Serotonin receptor 1a; Htr1b: Serotonin receptor 1b; SERT: Serotonin transporter; VMAT: Vesicular monoamine transporter; MAO: Monoamine oxidase.
Collapse
Affiliation(s)
- Ria Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| | - Swesha Chhabra
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Aditi Giri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Kajal Sherawat
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| |
Collapse
|
21
|
Wang J, Ware K, Bedolla A, Allgire E, Turcato FC, Weed M, Sah R, Luo Y. Disruption of Sonic Hedgehog Signaling Accelerates Age-Related Neurogenesis Decline and Abolishes Stroke-Induced Neurogenesis and Leads to Increased Anxiety Behavior in Stroke Mice. Transl Stroke Res 2022; 13:830-844. [PMID: 35146631 PMCID: PMC10114538 DOI: 10.1007/s12975-022-00994-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/10/2022] [Accepted: 01/31/2022] [Indexed: 02/03/2023]
Abstract
Sonic Hedgehog (SHH) signaling has a critical role in mediating developmental neurogenesis and has been implicated in adult subventricular (SVZ) neurogenesis. However, the precise role of Smoothened (SMO) receptor-mediated SHH signaling in adult neurogenesis during aging especially in hippocampal subgranular zone (SGZ) neurogenesis remains undefined. Additionally, our previous study showed that stimulation of SHH signaling post-stroke leads to increased neurogenesis and improved behavioral functions after stroke. However, it is not clear whether SHH signaling in neural stem cells (NSCs) is required for stroke-induced neurogenesis and functional recovery post-stroke. In this study, using conditional knockout (cKO) of SHH signaling receptor Smo gene in NSCs, we show a decreased neurogenesis at both SVZ and SGZ in young-adult mice and an accelerated depletion of neurogenic cells in the process of aging suggesting that SHH signaling is critical in maintaining neurogenesis during aging. Behavior studies revealed that compromised neurogenesis in Smo cKO mice leads to increased anxiety/depression-like behaviors without affecting general locomotor function or spatial and fear-related learning. Importantly, we also show that NSCs with a cKO of SHH signaling abolishes stroke-induced neurogenesis in Smo cKO mice. Compared to control mice, Smo cKO mice also show delayed motor function recovery and increased anxiety level after stroke. Our data highlights the essential role of Smo function in regulating adult neurogenesis and emotional behaviors during both aging and CNS injury such as stroke.
Collapse
Affiliation(s)
- Jiapeng Wang
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, 45267, USA
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Kierra Ware
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Alicia Bedolla
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Emily Allgire
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Flavia Correa Turcato
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Maxwell Weed
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Renu Sah
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, 45267, USA
- Cincinnati VA Medical Center, Cincinnati, OH, 45220, USA
| | - Yu Luo
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA.
| |
Collapse
|
22
|
Paro MR, Chakraborty AR, Angelo S, Nambiar S, Bulsara KR, Verma R. Molecular mediators of angiogenesis and neurogenesis after ischemic stroke. Rev Neurosci 2022; 34:425-442. [PMID: 36073599 DOI: 10.1515/revneuro-2022-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/22/2022] [Indexed: 11/15/2022]
Abstract
The mechanisms governing neurological and functional recovery after ischemic stroke are incompletely understood. Recent advances in knowledge of intrinsic repair processes of the CNS have so far translated into minimal improvement in outcomes for stroke victims. Better understanding of the processes underlying neurological recovery after stroke is necessary for development of novel therapeutic approaches. Angiogenesis and neurogenesis have emerged as central mechanisms of post-stroke recovery and potential targets for therapeutics. Frameworks have been developed for conceptualizing cerebral angiogenesis and neurogenesis at the tissue and cellular levels. These models highlight that angiogenesis and neurogenesis are linked to each other and to functional recovery. However, knowledge of the molecular framework linking angiogenesis and neurogenesis after stroke is limited. Studies of potential therapeutics typically focus on one mediator or pathway with minimal discussion of its role within these multifaceted biochemical processes. In this article, we briefly review the current understanding of the coupled processes of angiogenesis and neurogenesis after stroke. We then identify the molecular mediators and signaling pathways found in pre-clinical studies to upregulate both processes after stroke and contextualizes them within the current framework. This report thus contributes to a more-unified understanding of the molecular mediators governing angiogenesis and neurogenesis after stroke, which we hope will help guide the development of novel therapeutic approaches for stroke survivors.
Collapse
Affiliation(s)
- Mitch R Paro
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA.,Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Arijit R Chakraborty
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - Sophia Angelo
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - Shyam Nambiar
- University of Connecticut, 75 North Eagleville Rd, Storrs, CT 06269, USA
| | - Ketan R Bulsara
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA.,Division of Neurosurgery, University of Connecticut Health, 135 Dowling Way, Farmington, CT 06030, USA
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA.,Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| |
Collapse
|
23
|
Luo F, Wang J, Zhang Z, You Z, Bedolla A, Okwubido-Williams F, Huang LF, Silver J, Luo Y. Inhibition of CSPG receptor PTPσ promotes migration of newly born neuroblasts, axonal sprouting, and recovery from stroke. Cell Rep 2022; 40:111137. [PMID: 35905716 DOI: 10.1016/j.celrep.2022.111137] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/16/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
In addition to neuroprotective strategies, neuroregenerative processes could provide targets for stroke recovery. However, the upregulation of inhibitory chondroitin sulfate proteoglycans (CSPGs) impedes innate regenerative efforts. Here, we examine the regulatory role of PTPσ (a major proteoglycan receptor) in dampening post-stroke recovery. Use of a receptor modulatory peptide (ISP) or Ptprs gene deletion leads to increased neurite outgrowth and enhanced NSCs migration upon inhibitory CSPG substrates. Post-stroke ISP treatment results in increased axonal sprouting as well as neuroblast migration deeply into the lesion scar with a transcriptional signature reflective of repair. Lastly, peptide treatment post-stroke (initiated acutely or more chronically at 7 days) results in improved behavioral recovery in both motor and cognitive functions. Therefore, we propose that CSPGs induced by stroke play a predominant role in the regulation of neural repair and that blocking CSPG signaling pathways will lead to enhanced neurorepair and functional recovery in stroke.
Collapse
Affiliation(s)
- Fucheng Luo
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Jiapeng Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Zhen Zhang
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Zhen You
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alicia Bedolla
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - FearGod Okwubido-Williams
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - L Frank Huang
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yu Luo
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
24
|
Haggag MA, Salem AES, Elsherbini AM. Sustained Release In Situ Thermogelling Hydrogel of Cerebrolysin for Treatment of Facial Nerve Axotomy in Rats. J Oral Maxillofac Surg 2022; 80:949-959. [PMID: 35041809 DOI: 10.1016/j.joms.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE The most essential principle in managing facial nerve (FN) injury is proper diagnosis and early treatment. This study evaluated local application of different concentrations and injection intervals of Cerebrolysin hydrogel (CBLH) for facial nerve axotomy (FNA) treatment. We hypothesized that local application of CBLH may provide a sustained release of Cerebrolysin and enhance neural regeneration. METHODS The authors implemented a randomized, controlled, blinded animal study. The sample was composed of the right FN. Functionally, eye-blink reflex was evaluated 2 and 4 weeks postoperatively. All rats were euthanized after 4 weeks, and nerve regeneration was evaluated histopathologically and immunohistochemically (IHC) with antibody against neurofilament (anti-NF) and S100 proteins. Descriptive and correlation statistics were computed, and the P value was set at .05. RESULTS The sample was composed of 72 adult male rats equally allocated into 8 groups. Groups I and V served as control groups and were injected with phosphate buffered saline once and four times, respectively. Rest of the groups were injected with 5%, 10%, and 15% CBLH once in groups II, III, IV and weekly in groups VI, VII, and VIII. CBLH showed statistically significant FN regeneration by enhancing Schwann and axonal growth compared to control group especially with single injection of 10%, 15%, and 5% 4-time injections, where the P value was less than .001. Significant improvement of eye-blink reflex was correlated with structural improvement associated with CBLH. CONCLUSION Finally, CBLH enhanced nerve regeneration and rehabilitation after FNA in rats. Therefore, it could be considered as an alternative treatment of FNA. More experimental and clinical trials should be considered to detect the effectiveness of CBLH in neural regeneration.
Collapse
Affiliation(s)
- Mai Ahmed Haggag
- Assistant Professor, Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Mansoura University, Egypt
| | - Amgad El-Said Salem
- Lecturer, Pharmaceutics Department, Faculty of Pharmacy, Mansoura University, Egypt
| | - Amira M Elsherbini
- Assistant Professor, Oral Biology Department, Faculty of Dentistry, Mansoura University, Egypt.
| |
Collapse
|
25
|
Vicente-Acosta A, Giménez-Cassina A, Díaz-Nido J, Loria F. The smoothened agonist SAG reduces mitochondrial dysfunction and neurotoxicity of frataxin-deficient astrocytes. J Neuroinflammation 2022; 19:93. [PMID: 35413853 PMCID: PMC9006607 DOI: 10.1186/s12974-022-02442-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Friedreich's ataxia is a rare hereditary neurodegenerative disease caused by decreased levels of the mitochondrial protein frataxin. Similar to other neurodegenerative pathologies, previous studies suggested that astrocytes might contribute to the progression of the disease. To fully understand the mechanisms underlying neurodegeneration in Friedreich's ataxia, we investigated the reactivity status and functioning of cultured human astrocytes after frataxin depletion using an RNA interference-based approach and tested the effect of pharmacologically modulating the SHH pathway as a novel neuroprotective strategy. RESULTS We observed loss of cell viability, mitochondrial alterations, increased autophagy and lipid accumulation in cultured astrocytes upon frataxin depletion. Besides, frataxin-deficient cells show higher expression of several A1-reactivity markers and release of pro-inflammatory cytokines. Interestingly, most of these defects were prevented by chronically treating the cells with the smoothened agonist SAG. Furthermore, in vitro culture of neurons with conditioned medium from frataxin-deficient astrocytes results in a reduction of neuronal survival, neurite length and synapse formation. However, when frataxin-deficient astrocytes were chronically treated with SAG, we did not observe these alterations in neurons. CONCLUSIONS Our results demonstrate that the pharmacological activation of the SHH pathway could be used as a target to modulate astrocyte reactivity and neuron-glia interactions to prevent neurodegeneration in Friedreich's ataxia.
Collapse
Affiliation(s)
- Andrés Vicente-Acosta
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro, Segovia de Arana, Hospital Universitario Puerta de Hierro, Joaquín Rodrigo 1, Majadahonda, 28222 Madrid, Spain
- Program in Molecular Biosciences, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alfredo Giménez-Cassina
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain
| | - Javier Díaz-Nido
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro, Segovia de Arana, Hospital Universitario Puerta de Hierro, Joaquín Rodrigo 1, Majadahonda, 28222 Madrid, Spain
| | - Frida Loria
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Budapest 1, Alcorcón, 28922 Madrid, Spain
| |
Collapse
|
26
|
Role and Impact of Cerebrolysin for Ischemic Stroke Care. J Clin Med 2022; 11:jcm11051273. [PMID: 35268364 PMCID: PMC8911124 DOI: 10.3390/jcm11051273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 01/01/2023] Open
Abstract
Stroke is still a significant health problem that affects millions of people worldwide, as it is the second-leading cause of death and the third-leading cause of disability. Many changes have occurred in the treatment of acute ischemic stroke. Although the innovative concepts of neuroprotection and neurorecovery have been vigorously investigated in a substantial number of clinical studies in the past, only a few trials managed to increase the number of promising outcomes with regard to the multidimensional construct of brain protection and rehabilitation. In terms of pharmacological therapies with proven benefits in the post-ischemic process, drugs with neurorestorative properties are thought to be effective in both the acute and chronic phases of stroke. One significant example is Cerebrolysin, a combination of amino acids and peptides that mimic the biological functions of neurotrophic factors, which has been shown to improve outcomes after ischemic stroke, while preserving a promising safety profile. The purpose of this paper is to offer an overview on the role and impact of Cerebrolysin for ischemic stroke care, by touching on various aspects, from its complex, multimodal and pleiotropic mechanism of action, to its efficacy and safety, as well as cost effectiveness.
Collapse
|
27
|
Burton DF, Boa-Amponsem OM, Dixon MS, Hopkins MJ, Herbin TA, Toney S, Tarpley M, Rodriguez BV, Fish EW, Parnell SE, Cole GJ, Williams KP. Pharmacological activation of the Sonic hedgehog pathway with a Smoothened small molecule agonist ameliorates the severity of alcohol-induced morphological and behavioral birth defects in a zebrafish model of fetal alcohol spectrum disorder. J Neurosci Res 2022; 100:1585-1601. [PMID: 35014067 PMCID: PMC9271529 DOI: 10.1002/jnr.25008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 12/17/2022]
Abstract
Ethanol exposure during the early stages of embryonic development can lead to a range of morphological and behavioral differences termed fetal alcohol spectrum disorders (FASDs). In a zebrafish model, we have shown that acute ethanol exposure at 8-10 hr postfertilization (hpf), a critical time of development, produces birth defects similar to those clinically characterized in FASD. Dysregulation of the Sonic hedgehog (Shh) pathway has been implicated as a molecular basis for many of the birth defects caused by prenatal alcohol exposure. We observed in zebrafish embryos that shh expression was significantly decreased by ethanol exposure at 8-10 hpf, while smo expression was much less affected. Treatment of zebrafish embryos with SAG or purmorphamine, small molecule Smoothened agonists that activate Shh signaling, ameliorated the severity of ethanol-induced developmental malformations including altered eye size and midline brain development. Furthermore, this rescue effect of Smo activation was dose dependent and occurred primarily when treatment was given after ethanol exposure. Markers of Shh signaling (gli1/2) and eye development (pax6a) were restored in embryos treated with SAG post-ethanol exposure. Since embryonic ethanol exposure has been shown to produce later-life neurobehavioral impairments, juvenile zebrafish were examined in the novel tank diving test. Our results further demonstrated that in zebrafish embryos exposed to ethanol, SAG treatment was able to mitigate long-term neurodevelopmental impairments related to anxiety and risk-taking behavior. Our results indicate that pharmacological activation of the Shh pathway at specific developmental timing markedly diminishes the severity of alcohol-induced birth defects.
Collapse
Affiliation(s)
- Derek F Burton
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA
| | - Oswald M Boa-Amponsem
- Integrated Biosciences PhD Program, North Carolina Central University, Durham, North Carolina, USA.,Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, USA
| | - Maria S Dixon
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA
| | - Michael J Hopkins
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Te-Andre Herbin
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Shiquita Toney
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Michael Tarpley
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA
| | - Blanca V Rodriguez
- Department of Biochemistry, Duke University, Durham, North Carolina, USA
| | - Eric W Fish
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Gregory J Cole
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, USA.,Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Kevin P Williams
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA.,Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| |
Collapse
|
28
|
Bedolla A, Taranov A, Luo F, Wang J, Turcato F, Fugate EM, Greig NH, Lindquist DM, Crone SA, Goto J, Luo Y. Diphtheria toxin induced but not CSF1R inhibitor mediated microglia ablation model leads to the loss of CSF/ventricular spaces in vivo that is independent of cytokine upregulation. J Neuroinflammation 2022; 19:3. [PMID: 34983562 PMCID: PMC8728932 DOI: 10.1186/s12974-021-02367-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Background Two recently developed novel rodent models have been reported to ablate microglia, either by genetically targeting microglia (via Cx3cr1-creER: iDTR + Dtx) or through pharmacologically targeting the CSF1R receptor with its inhibitor (PLX5622). Both models have been widely used in recent years to define essential functions of microglia and have led to high impact studies that have moved the field forward. Methods Using either Cx3cr1-iDTR mice in combination with Dtx or via the PLX5622 diet to pharmacologically ablate microglia, we compared the two models via MRI and histology to study the general anatomy of the brain and the CSF/ventricular systems. Additionally, we analyzed the cytokine profile in both microglia ablation models. Results We discovered that the genetic ablation (Cx3cr1-iDTR + Dtx), but not the pharmacological microglia ablation (PLX5622), displays a surprisingly rapid pathological condition in the brain represented by loss of CSF/ventricles without brain parenchymal swelling. This phenotype was observed both in MRI and histological analysis. To our surprise, we discovered that the iDTR allele alone leads to the loss of CSF/ventricles phenotype following diphtheria toxin (Dtx) treatment independent of cre expression. To examine the underlying mechanism for the loss of CSF in the Cx3cr1-iDTR ablation and iDTR models, we additionally investigated the cytokine profile in the Cx3cr1-iDTR + Dtx, iDTR + Dtx and the PLX models. We found increases of multiple cytokines in the Cx3cr1-iDTR + Dtx but not in the pharmacological ablation model nor the iDTR + Dtx mouse brains at the time of CSF loss (3 days after the first Dtx injection). This result suggests that the upregulation of cytokines is not the cause of the loss of CSF, which is supported by our data indicating that brain parenchyma swelling, or edema are not observed in the Cx3cr1-iDTR + Dtx microglia ablation model. Additionally, pharmacological inhibition of the KC/CXCR2 pathway (the most upregulated cytokine in the Cx3cr1-iDTR + Dtx model) did not resolve the CSF/ventricular loss phenotype in the genetic microglia ablation model. Instead, both the Cx3cr1-iDTR + Dtx ablation and iDTR + Dtx models showed increased activated IBA1 + cells in the choroid plexus (CP), suggesting that CP-related pathology might be the contributing factor for the observed CSF/ventricular shrinkage phenotype. Conclusions Our data, for the first time, reveal a robust and global CSF/ventricular space shrinkage pathology in the Cx3cr1-iDTR genetic ablation model caused by iDTR allele, but not in the PLX5622 ablation model, and suggest that this pathology is not due to brain edema formation but to CP related pathology. Given the wide utilization of the iDTR allele and the Cx3cr1-iDTR model, it is crucial to fully characterize this pathology to understand the underlying causal mechanisms. Specifically, caution is needed when utilizing this model to interpret subtle neurologic functional changes that are thought to be mediated by microglia but could, instead, be due to CSF/ventricular loss in the genetic ablation model.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Aleksandr Taranov
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Fucheng Luo
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jiapeng Wang
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Flavia Turcato
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Elizabeth M Fugate
- Imaging Research Center, Cincinnati Children's Hospital Medical Center, Department of Radiology, University of Cincinnati, Cincinnati, USA
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, USA
| | - Diana M Lindquist
- Imaging Research Center, Cincinnati Children's Hospital Medical Center, Department of Radiology, University of Cincinnati, Cincinnati, USA
| | - Steven A Crone
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Department of Neurosurgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - June Goto
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Department of Neurosurgery, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA
| | - Yu Luo
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
29
|
Nguyen V, Chavali M, Larpthaveesarp A, Kodali S, Gonzalez G, Franklin RJM, Rowitch DH, Gonzalez F. Neuroprotective effects of Sonic hedgehog agonist SAG in a rat model of neonatal stroke. Pediatr Res 2021; 90:1161-1170. [PMID: 33654279 PMCID: PMC8410885 DOI: 10.1038/s41390-021-01408-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 12/04/2022]
Abstract
BACKGROUND Neonatal stroke affects 1 in 2800 live births and is a major cause of neurological injury. The Sonic hedgehog (Shh) signaling pathway is critical for central nervous system (CNS) development and has neuroprotective and reparative effects in different CNS injury models. Previous studies have demonstrated beneficial effects of small molecule Shh-Smoothened agonist (SAG) against neonatal cerebellar injury and it improves Down syndrome-related brain structural deficits in mice. Here we investigated SAG neuroprotection in rat models of neonatal ischemia-reperfusion (stroke) and adult focal white matter injury. METHODS We used transient middle cerebral artery occlusion at P10 and ethidium bromide (EB) injection in adult rats to induce damage. Following surgery and SAG or vehicle treatment, we analyzed tissue loss, cell proliferation and fate, and behavioral outcome. RESULTS We report that a single dose of SAG administered following neonatal stroke preserved brain volume, reduced gliosis, enhanced oligodendrocyte progenitor cell (OPC) and EC proliferation, and resulted in long-term cognitive improvement. Single-dose SAG also promoted proliferation of OPCs following focal demyelination in the adult rat. CONCLUSIONS These findings indicate benefit of one-time SAG treatment post insult in reducing brain injury and improving behavioral outcome after experimental neonatal stroke. IMPACT A one-time dose of small molecule Sonic hedgehog agonist protected against neonatal stroke and improved long-term behavioral outcomes in a rat model. This study extends the use of Sonic hedgehog in treating developing brain injury, previously shown in animal models of Down syndrome and cerebellar injury. Sonic hedgehog agonist is one of the most promising therapies in treating neonatal stroke thanks to its safety profile and low dosage.
Collapse
Affiliation(s)
- Vien Nguyen
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Eli and Edyth Broad Center for Stem Cell Research and Regenerative Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Manideep Chavali
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Eli and Edyth Broad Center for Stem Cell Research and Regenerative Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Amara Larpthaveesarp
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Srikirti Kodali
- Jeffrey Cheah Biomedical Centre, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Ginez Gonzalez
- Jeffrey Cheah Biomedical Centre, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Robin J M Franklin
- Jeffrey Cheah Biomedical Centre, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - David H Rowitch
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
- Eli and Edyth Broad Center for Stem Cell Research and Regenerative Medicine, University of California San Francisco, San Francisco, CA, USA.
- Jeffrey Cheah Biomedical Centre, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| | - Fernando Gonzalez
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
30
|
Cucu I, Nicolescu MI. A Synopsis of Signaling Crosstalk of Pericytes and Endothelial Cells in Salivary Gland. Dent J (Basel) 2021; 9:dj9120144. [PMID: 34940041 PMCID: PMC8700478 DOI: 10.3390/dj9120144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
The salivary gland (SG) microvasculature constitutes a dynamic cellular organization instrumental to preserving tissue stability and homeostasis. The interplay between pericytes (PCs) and endothelial cells (ECs) culminates as a key ingredient that coordinates the development, maturation, and integrity of vessel building blocks. PCs, as a variety of mesenchymal stem cells, enthrall in the field of regenerative medicine, supporting the notion of regeneration and repair. PC-EC interconnections are pivotal in the kinetic and intricate process of angiogenesis during both embryological and post-natal development. The disruption of this complex interlinkage corresponds to SG pathogenesis, including inflammation, autoimmune disorders (Sjögren’s syndrome), and tumorigenesis. Here, we provided a global portrayal of major signaling pathways between PCs and ECs that cooperate to enhance vascular steadiness through the synergistic interchange. Additionally, we delineated how the crosstalk among molecular networks affiliate to contribute to a malignant context. Additionally, within SG microarchitecture, telocytes and myoepithelial cells assemble a labyrinthine companionship, which together with PCs appear to synchronize the regenerative potential of parenchymal constituents. By underscoring the intricacy of signaling cascades within cellular latticework, this review sketched a perceptive basis for target-selective drugs to safeguard SG function.
Collapse
Affiliation(s)
- Ioana Cucu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mihnea Ioan Nicolescu
- Division of Histology, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Laboratory of Radiobiology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
- Correspondence:
| |
Collapse
|
31
|
Antonelli F, Casciati A, Belles M, Serra N, Linares-Vidal MV, Marino C, Mancuso M, Pazzaglia S. Long-Term Effects of Ionizing Radiation on the Hippocampus: Linking Effects of the Sonic Hedgehog Pathway Activation with Radiation Response. Int J Mol Sci 2021; 22:ijms222212605. [PMID: 34830484 PMCID: PMC8624704 DOI: 10.3390/ijms222212605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 12/29/2022] Open
Abstract
Radiation therapy represents one of the primary treatment modalities for primary and metastatic brain tumors. Although recent advances in radiation techniques, that allow the delivery of higher radiation doses to the target volume, reduce the toxicity to normal tissues, long-term neurocognitive decline is still a detrimental factor significantly affecting quality of life, particularly in pediatric patients. This imposes the need for the development of prevention strategies. Based on recent evidence, showing that manipulation of the Shh pathway carries therapeutic potential for brain repair and functional recovery after injury, here we evaluate how radiation-induced hippocampal alterations are modulated by the constitutive activation of the Shh signaling pathway in Patched 1 heterozygous mice (Ptch1+/-). Our results show, for the first time, an overall protective effect of constitutive Shh pathway activation on hippocampal radiation injury. This activation, through modulation of the proneural gene network, leads to a long-term reduction of hippocampal deficits in the stem cell and new neuron compartments and to the mitigation of radio-induced astrogliosis, despite some behavioral alterations still being detected in Ptch1+/- mice. A better understanding of the pathogenic mechanisms responsible for the neural decline following irradiation is essential for identifying prevention measures to contain the harmful consequences of irradiation. Our data have important translational implications as they suggest a role for Shh pathway manipulation to provide the therapeutic possibility of improving brain repair and functional recovery after radio-induced injury.
Collapse
Affiliation(s)
- Francesca Antonelli
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
- Correspondence: (F.A.); (S.P.)
| | - Arianna Casciati
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
| | - Montserrat Belles
- Physiology Unit, School of Medicine, Rovira I Virgili University (URV), 43007 Reus, Spain; (M.B.); (N.S.); (M.V.L.-V.)
| | - Noemi Serra
- Physiology Unit, School of Medicine, Rovira I Virgili University (URV), 43007 Reus, Spain; (M.B.); (N.S.); (M.V.L.-V.)
| | - Maria Victoria Linares-Vidal
- Physiology Unit, School of Medicine, Rovira I Virgili University (URV), 43007 Reus, Spain; (M.B.); (N.S.); (M.V.L.-V.)
| | - Carmela Marino
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
| | - Simonetta Pazzaglia
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
- Correspondence: (F.A.); (S.P.)
| |
Collapse
|
32
|
Wang Y, Lu S, Chen Y, Li L, Li X, Qu Z, Huang J, Fan L, Yuan C, Song N, Zhang J, Xu W, Yang S, Wang Y. Smoothened is a therapeutic target for reducing glutamate toxicity in ischemic stroke. Sci Transl Med 2021; 13:eaba3444. [PMID: 34516830 DOI: 10.1126/scitranslmed.aba3444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yuqing Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China.,Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, 200031 Shanghai, China
| | - Shanshan Lu
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Yifei Chen
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Liang Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Xia Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Zhongwei Qu
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, 200031 Shanghai, China
| | - Junbo Huang
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, 200031 Shanghai, China
| | - Liu Fan
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, 200031 Shanghai, China
| | - Chao Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Nan Song
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Jun Zhang
- Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Wendong Xu
- Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Shenglian Yang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Yizheng Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China.,Huashan Hospital, Fudan University, 200040 Shanghai, China
| |
Collapse
|
33
|
Zhang Q, Dai J, Song Z, Guo Y, Deng S, Yu Y, Li T, Zhang Y. Anti-Inflammatory Dipeptide, a Metabolite from Ambioba Secretion, Protects Cerebral Ischemia Injury by Blocking Apoptosis Via p-JNK/Bax Pathway. Front Pharmacol 2021; 12:689007. [PMID: 34220513 PMCID: PMC8249563 DOI: 10.3389/fphar.2021.689007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/03/2021] [Indexed: 12/03/2022] Open
Abstract
MQ (l-methionyl-l-glutamic acid), anti-inflammatory dipeptide, is one of the metabolites of monocyte locomotion inhibitory factor, a thermostable pentapeptide secreted by Entamoeba histolytica. Monocyte locomotion inhibitory factor injection has been approved as an investigational drug for the potential neural protection in acute ischemic stroke. This study further investigated the neuroprotective effect of MQ in ischemic brain damage. Ischemia-reperfusion injury of the brain was induced in the rat model by middle cerebral artery occlusion. 2,3,5-triphenyltetrazolium chloride staining assay was used to measure cerebral infarction areas in rats. Laser Doppler measurement instrument was used to detect blood flow changes in the rat model. Nissl staining and NeuN staining were utilized to observe the numbers and structures of neuron cells, and the pathological changes in the brain tissues were examined by hematoxylin–eosin staining. Terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling (TUNEL) staining was used to assess cell apoptosis. The changes in oxidative stress indexes, superoxide dismutase and malondialdehyde (MDA), were measured in serum. Methyl thiazolyl tetrazolium was used to measure the survival rates of PC12 cells. Flow cytometry assessed the apoptosis rates and the levels of reactive oxygen species. Real-time PCR was used to evaluate the mRNA expression levels, and Western blotting was used to analyze the changes in protein levels of p-JNK, Bax, cleaved Caspase3. We revealed that MQ improved neurobehavior, decreased cerebral infarction areas, altered blood flow volume, and the morphology of the cortex and hippocampus. On the other hand, it decreased the apoptosis of cortical neurons and the levels of MDA, and increased the levels of superoxide dismutase. In vitro studies demonstrated that MQ enhanced the cell survival rates and decreased the levels of reactive oxygen species. Compared to the oxygen-glucose deprivation/reperfusion group, the protein and mRNA expressions of p-JNK, Bax, cleaved Caspase3 was decreased significantly. These findings suggested that MQ exerts a neuroprotective effect in cerebral ischemia by blocking apoptosis via the p-JNK/Bax pathway.
Collapse
Affiliation(s)
- Qian Zhang
- School of Medicine, Shanghai University, Shanghai, China.,College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Jinwei Dai
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhibing Song
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Yuchen Guo
- College of Pharmacology, Anhui University of Chinese Medicine, Hefei, China
| | - Shanshan Deng
- School of Medicine, Shanghai University, Shanghai, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shanghai, China
| | - Tiejun Li
- School of Medicine, Shanghai University, Shanghai, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
34
|
Zhu W, Romano KA, Li L, Buffa JA, Sangwan N, Prakash P, Tittle AN, Li XS, Fu X, Androjna C, DiDonato AJ, Brinson K, Trapp BD, Fischbach MA, Rey FE, Hajjar AM, DiDonato JA, Hazen SL. Gut microbes impact stroke severity via the trimethylamine N-oxide pathway. Cell Host Microbe 2021; 29:1199-1208.e5. [PMID: 34139173 DOI: 10.1016/j.chom.2021.05.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022]
Abstract
Clinical studies have demonstrated associations between circulating levels of the gut-microbiota-derived metabolite trimethylamine-N-oxide (TMAO) and stroke incident risk. However, a causal role of gut microbes in stroke has not yet been demonstrated. Herein we show that gut microbes, through dietary choline and TMAO generation, directly impact cerebral infarct size and adverse outcomes following stroke. Fecal microbial transplantation from low- versus high-TMAO-producing human subjects into germ-free mice shows that both TMAO generation and stroke severity are transmissible traits. Furthermore, employing multiple murine stroke models and transplantation of defined microbial communities with genetically engineered human commensals into germ-free mice, we demonstrate that the microbial cutC gene (an enzymatic source of choline-to-TMA transformation) is sufficient to transmit TMA/TMAO production, heighten cerebral infarct size, and lead to functional impairment. We thus reveal that gut microbiota in general, specifically the metaorganismal TMAO pathway, directly contributes to stroke severity.
Collapse
Affiliation(s)
- Weifei Zhu
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Kymberleigh A Romano
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lin Li
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jennifer A Buffa
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Prem Prakash
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Aaron N Tittle
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xiaoming Fu
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Charlie Androjna
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anthony J DiDonato
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kimberly Brinson
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Bruce D Trapp
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael A Fischbach
- Department of Bioengineering and ChEM-H, Stanford University, Stanford, CA, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Adeline M Hajjar
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Joseph A DiDonato
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA; Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
35
|
Yu P, Wang L, Tang F, Guo S, Liao H, Fan C, Yang Q. Resveratrol-mediated neurorestoration after cerebral ischemic injury - Sonic Hedgehog signaling pathway. Life Sci 2021; 280:119715. [PMID: 34116113 DOI: 10.1016/j.lfs.2021.119715] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/11/2021] [Accepted: 05/22/2021] [Indexed: 11/16/2022]
Abstract
AIMS Resveratrol pretreatment can decrease ischemic cerebral injury and enhance proliferation of neural stem cells via mediation of Sonic Hedgehog signaling. However, it is relatively little known about whether neurorestorative effects of resveratrol are mediated by Shh signaling in ischemic cerebral injury. The present study tests whether the Shh signaling pathway mediates resveratrol to promote neurorestoration of ischemic cerebral injury. MATERIALS AND METHODS Rats or neurons before middle cerebral artery occlusion/reperfusion (MCAO/R) or oxygen-glucose deprivation/reoxygenation (OGD/R) injury were pretreated with resveratrol. Immunohistochemistry is used to be determined BrdU+/DCX+, BrdU+/Nestin+ and BrdU+/NG2+ cell (markers of new proliferated neural stem/progenitor and oligodendrocyte precursor cell, respectively), BrdU+/MAP2+ and BrdU+/CNPase+ cell (markers of new mature neuron and oligodendrocyte, respectively), BrdU+/TUNEL+ cell (marker of apoptosis for new proliferated cell), SY, NF200, Iba-1 and GFAP (markers of synaptogenesis, axon, microglia and astrocyte, respectively). Shh and Gli-1 mRNAs were detected by RT-PCR assay. Iba-1, GFAP, Shh and Gli-1 proteins were detected by Western blot. KEY FINDINGS Resveratrol pretreatment significantly reduced neurological deficit scores, promoted proliferation, differentiation, migration and survival of neural stem/progenitor and oligodendrocyte precursor cells, inhibited astrocyte and microglia activation, strengthened synaptophysin and NF200 expression, at the same time, promoted neurite outgrowth of neurons. Meanwhile, expression levels of Shh and Gli-1 proteins were significantly increased and Gli-1 translocated into the nucleus. However, cyclopamine, a Smo inhibitor, canceled the above effects of resveratrol. CONCLUSIONS It may be mediated, at least partly, by the Shh signaling pathway that resveratrol pretreament promote neurorestoration of ischemic cerebral injury.
Collapse
Affiliation(s)
- Pingping Yu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Physical Examination Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fanren Tang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuang Guo
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongyan Liao
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Cengceng Fan
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Yang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
36
|
Yang H, Luo Y, Hu H, Yang S, Li Y, Jin H, Chen S, He Q, Hong C, Wu J, Wan Y, Li M, Li Z, Yang X, Su Y, Zhou Y, Hu B. pH-Sensitive, Cerebral Vasculature-Targeting Hydroxyethyl Starch Functionalized Nanoparticles for Improved Angiogenesis and Neurological Function Recovery in Ischemic Stroke. Adv Healthc Mater 2021; 10:e2100028. [PMID: 34028998 DOI: 10.1002/adhm.202100028] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/16/2021] [Indexed: 01/17/2023]
Abstract
Angiogenesis, an essential restorative process following ischemia, is a promising therapeutic approach to improve neurological deficits. However, overcoming the blood-brain barrier (BBB) and effective drug enrichment are challenges for conventional drug delivery methods, which has limited the development of treatment strategies. Herein, a dual-targeted therapeutic strategy is reported to enable pH-sensitive drug release and allow cerebral ischemia targeting to improve stroke therapeutic efficacy. Targeted delivery is achieved by surface conjugation of Pro-His-Ser-Arg-Asn (PHSRN) peptides, which binds to integrin α5 β1 enriched in the cerebral vasculature of ischemic tissue. Subsequently, smoothened agonist (SAG), an activator of sonic hedgehog (Shh) signaling, is coupled to PHSRN-HES by pH-dependent electrostatic adsorption. SAG@PHSRN-HES nanoparticles can sensitively release more SAG in the acidic environment of ischemic brain tissue. More importantly, SAG@PHSRN-HES exerts the synergistic mechanisms of PHSRN and SAG to promote angiogenesis and BBB integrity, thus improving neuroplasticity and neurological function recovery. This study proposes a new approach to improve the delivery of medications in the ischemic brain. Dual-targeted therapeutic strategies have excellent potential to treat patients suffering from cerebral infarction.
Collapse
Affiliation(s)
- Hang Yang
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Yan Luo
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Hang Hu
- School of Pharmacy Changzhou University Changzhou 213164 P. R. China
| | - Sibo Yang
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Yanan Li
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Huijuan Jin
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Shengcai Chen
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Quanwei He
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Candong Hong
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Jiehong Wu
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Yan Wan
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Man Li
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology Wuhan 430074 P. R. China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology Wuhan 430074 P. R. China
| | - Ying Su
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Yifan Zhou
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Bo Hu
- Department of Neurology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| |
Collapse
|
37
|
Liu W, Ge X, Zhou Z, Jiang D, Rong Y, Wang J, Ji C, Fan J, Yin G, Cai W. Deubiquitinase USP18 regulates reactive astrogliosis by stabilizing SOX9. Glia 2021; 69:1782-1798. [PMID: 33694203 DOI: 10.1002/glia.23992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 02/24/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022]
Abstract
Reactive astrogliosis is a pathological feature of spinal cord injury (SCI). The ubiquitin-proteasome system plays a crucial role in maintaining protein homeostasis and has been widely studied in neuroscience. Little, however, is known about the underlying function of deubiquitinating enzymes in reactive astrogliosis following SCI. Here, we found that ubiquitin-specific protease 18 (USP18) was significantly upregulated in astrocytes following scratch injury, and in the injured spinal cord in mice. Knockdown of USP18 in vitro and conditional knockout of USP18 in astrocytes (USP18 CKO) in vivo significantly attenuated reactive astrogliosis. In mice, this led to widespread inflammation and poor functional recovery following SCI. In contrast, overexpression of USP18 in mice injected with adeno-associated virus (AAV)-USP18 had beneficial effects following SCI. We showed that USP18 binds, deubiquitinates, and thus, stabilizes SRY-box transcription factor 9 (SOX9), thereby regulating reactive astrogliosis. We also showed that the Hedgehog (Hh) signaling pathway induces expression of USP18 through Gli2-mediated transcriptional activation after SCI. Administration of the Hh pathway activator SAG significantly increased reactive astrogliosis, reduced lesion area and promoted functional recovery in mice following SCI. Our results demonstrate that USP18 positively regulates reactive astrogliosis by stabilizing SOX9 and identify USP18 as a promising target for the treatment of SCI.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuhui Ge
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheng Zhou
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongdong Jiang
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuluo Rong
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaxing Wang
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chengyue Ji
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin Fan
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoyong Yin
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weihua Cai
- Department of Orthopaedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
38
|
Feng L, Dou C, Xia Y, Li B, Zhao M, Yu P, Zheng Y, El-Toni AM, Atta NF, Galal A, Cheng Y, Cai X, Wang Y, Zhang F. Neutrophil-like Cell-Membrane-Coated Nanozyme Therapy for Ischemic Brain Damage and Long-Term Neurological Functional Recovery. ACS NANO 2021; 15:2263-2280. [PMID: 33426885 DOI: 10.1021/acsnano.0c07973] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Oxidative stress and a series of excessive inflammatory responses are major obstacles for neurological functional recovery after ischemic stroke. Effective noninvasive anti-inflammatory therapies are urgently needed. However, unsatisfactory therapeutic efficacy of current drugs and inadequate drug delivery to the damaged brain are major problems. Nanozymes with robust anti-inflammatory and antioxidative stress properties possess therapeutic possibility for ischemic stroke. However, insufficiency of nanozyme accumulation in the ischemic brain by noninvasive administration hindered their application. Herein, we report a neutrophil-like cell-membrane-coated mesoporous Prussian blue nanozyme (MPBzyme@NCM) to realize noninvasive active-targeting therapy for ischemic stroke by improving the delivery of a nanozyme to the damaged brain based on the innate connection between inflamed brain microvascular endothelial cells and neutrophils after stroke. The long-term in vivo therapeutic efficacy of MPBzyme@NCM for ischemic stroke was illustrated in detail after being delivered into the damaged brain and uptake by microglia. Moreover, the detailed mechanism of ischemic stroke therapy via MPBzyme@NCM uptake by microglia was further studied, including microglia polarization toward M2, reduced recruitment of neutrophils, decreased apoptosis of neurons, and proliferation of neural stem cells, neuronal precursors, and neurons. This strategy may provide an applicative perspective for nanozyme therapy in brain diseases.
Collapse
Affiliation(s)
- Lishuai Feng
- Department of Radiology, Department of Ultrasound in Medicine, and Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Chaoran Dou
- Department of Radiology, Department of Ultrasound in Medicine, and Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Yuguo Xia
- Department of Radiology, Department of Ultrasound in Medicine, and Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Benhao Li
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, People's Republic of China
| | - Mengyao Zhao
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, People's Republic of China
| | - Peng Yu
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, People's Republic of China
| | - Yuanyi Zheng
- Department of Radiology, Department of Ultrasound in Medicine, and Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Ahmed Mohamed El-Toni
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Central Metallurgical Research and Development Institute (CMRDI), Helwan, 11421 Cairo, Egypt
| | - Nada Farouk Atta
- Department of Chemistry, Cairo University, Giza, 12613 Cairo, Egypt
| | - Ahmed Galal
- Department of Chemistry, Cairo University, Giza, 12613 Cairo, Egypt
| | - Yingsheng Cheng
- Department of Radiology, Department of Ultrasound in Medicine, and Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Xiaojun Cai
- Department of Radiology, Department of Ultrasound in Medicine, and Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Yan Wang
- Department of Radiology, Department of Ultrasound in Medicine, and Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Fan Zhang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, People's Republic of China
| |
Collapse
|
39
|
Pringle AK, Solomon E, Coles BJ, Desousa BR, Shtaya A, Gajavelli S, Dabab N, Zaben MJ, Bulters DO, Bullock MR, Ahmed AI. Sonic Hedgehog Signaling Promotes Peri-Lesion Cell Proliferation and Functional Improvement after Cortical Contusion Injury. Neurotrauma Rep 2021; 2:27-38. [PMID: 33748811 PMCID: PMC7962778 DOI: 10.1089/neur.2020.0016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability globally. No drug treatments are available, so interest has turned to endogenous neural stem cells (NSCs) as alternative strategies for treatment. We hypothesized that regulation of cell proliferation through modulation of the sonic hedgehog pathway, a key NSC regulatory pathway, could lead to functional improvement. We assessed sonic hedgehog (Shh) protein levels in the cerebrospinal fluid (CSF) of patients with TBI. Using the cortical contusion injury (CCI) model in rodents, we used pharmacological modulators of Shh signaling to assess cell proliferation within the injured cortex using the marker 5-Ethynyl-2’-deoxyuridine (EdU); 50mg/mL. The phenotype of proliferating cells was determined and quantified. Motor function was assessed using the rotarod test. In patients with TBI there is a reduction of Shh protein in CSF compared with control patients. In rodents, following a severe CCI, quiescent cells become activated. Pharmacologically modulating the Shh signaling pathway leads to changes in the number of newly proliferating injury-induced cells. Upregulation of Shh signaling with Smoothened agonist (SAG) results in an increase of newly proliferating cells expressing glial fibrillary acidic protein (GFAP), whereas the Shh signaling inhibitor cyclopamine leads to a reduction. Some cells expressed doublecortin (DCX) but did not mature into neurons. The SAG-induced increase in proliferation is associated with improved recovery of motor function. Localized restoration of Shh in the injured rodent brain, via increased Shh signaling, has the potential to sustain endogenous cell proliferation and the mitigation of TBI-induced motor deficits albeit without the neuronal differentiation.
Collapse
Affiliation(s)
- Ashley K Pringle
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Elshadaie Solomon
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Benjamin J Coles
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Brandon R Desousa
- Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Anan Shtaya
- Neurosciences Research Centre, St. George's, University of London, London, United Kingdom
| | - Shyam Gajavelli
- Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Nedal Dabab
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Malik J Zaben
- Neuroscience and Mental Health Research Institute, University of Cardiff, Cardiff, Wales, United Kingdom
| | - Diederik O Bulters
- Wessex Neurological Centre, University Hospitals Southampton NHS Trust, Southampton, United Kingdom
| | - M Ross Bullock
- Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Aminul I Ahmed
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,Brain Repair and Rehabilitation, Institute of Neurology, London, United Kingdom
| |
Collapse
|
40
|
Rahi S, Mehan S. Understanding Abnormal SMO-SHH Signaling in Autism Spectrum Disorder: Potential Drug Target and Therapeutic Goals. Cell Mol Neurobiol 2020; 42:931-953. [PMID: 33206287 DOI: 10.1007/s10571-020-01010-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Autism is a multifactorial neurodevelopmental condition; it demonstrates some main characteristics, such as impaired social relationships and increased repetitive behavior. The initiation of autism spectrum disorder is mostly triggered during brain development by the deregulation of signaling pathways. Sonic hedgehog (SHH) signaling is one such mechanism that influences neurogenesis and neural processes during the development of the central nervous system. SMO-SHH signaling is also an important part of a broad variety of neurological processes, including neuronal cell differentiation, proliferation, and survival. Dysregulation of SMO-SHH signaling leads to many physiological changes that lead to neurological disorders such as ASD and contribute to cognitive decline. The aberrant downregulation of SMO-SHH signals contributes to the proteolytic cleavage of GLI (glioma-associated homolog) into GLI3 (repressor), which increases oxidative stress, neuronal excitotoxicity, neuroinflammation, and apoptosis by suppressing target gene expression. We outlined in this review that SMO-SHH deregulation plays a crucial role in the pathogenesis of autism and addresses the current status of SMO-SHH pathway modulators. Additionally, a greater understanding of the SHH signaling pathway is an effort to improve successful treatment for autism and other neurological disorders.
Collapse
Affiliation(s)
- Saloni Rahi
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
41
|
Xiang X, Wang L, Zhou L, Chen Y, Xia H. Metformin upregulates the expression of Gli1 in vascular endothelial cells in hyperoxia-exposed neonatal mice. Am J Transl Res 2020; 12:6092-6106. [PMID: 33194016 PMCID: PMC7653632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by arrested alveolar and vascular development in premature infants. Metformin protects against the cardiovascular impairment induced by diabetes. The aim of this study was to investigate whether metformin could also enhance pulmonary vascular development in hyperoxic neonatal mice and investigate possible mechanisms involved. C57BL/6J newborn mice were randomly assigned to either of two groups - the room air group or the hyperoxia group - within 12 h postnatally. The mice were subcutaneously injected with metformin (100 mg/kg) or saline for 14 days. Lung morphology and PECAM-1 (CD31) expression in the lung were evaluated at postnatal days 7 and 14. Ki-67 and Gli1 expression in vascular endothelial cells was evaluated at postnatal day 14 by immunofluorescence staining. Flow cytometry (FCM) was also used to analyze Gli1 expression. Human umbilical vein endothelial cell (HUVECs) were used to investigate the role of metformin in vascular proliferation and tubular formation under 90% oxygen in vitro by cell counting Kti-8 (CCK8) assays and tube formation assays. Exposure to hyperoxia resulted in impaired lung development in newborn mice. Metformin enhanced the terminal airspace and radial alveolar count in newborn mice thus exposed. Immunohistochemistry staining and western blot assays revealed that metformin enhanced the expression of CD31 in hyperoxia-exposed newborn mice. Immunofluorescence staining showed that metformin enhanced the expression of Ki-67 in vascular endothelial cells. Furthermore, both immunofluorescence staining and FCM demonstrated that metformin increased Gli1 expression in vascular endothelial cells. Additionally, cell counting Kit-8 (CCK8) and viability assays of HUVECs in vitro both indicated that metformin improved the vascular proliferation and tube formation of HUVECs under 90% oxygen. These results indicated that metformin enhanced lung vascular development and upregulated the expression of Gli1 in the pulmonary vascular endothelial cells in hyperoxic neonatal mice.
Collapse
Affiliation(s)
- Xiaowen Xiang
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Li Wang
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Lin Zhou
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Yanru Chen
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Hongping Xia
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| |
Collapse
|
42
|
Aziz NS, Yusop N, Ahmad A. Importance of Stem Cell Migration and Angiogenesis Study for Regenerative Cell-based Therapy: A Review. Curr Stem Cell Res Ther 2020; 15:284-299. [DOI: 10.2174/1574888x15666200127145923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/01/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022]
Abstract
Stem cells play an essential role in maintaining homeostasis, as well as participating in new
tissue regeneration. Over the past 20 years, a great deal of effort has been made to investigate the behaviour
of stem cells to enable their potential use in regenerative medicine. However, a variety of biological
characteristics are known to exist among the different types of stem cells due to variations in
the methodological approach, formulation of cell culture medium, isolation protocol and cellular
niches, as well as species variation. In recent years, cell-based therapy has emerged as one of the advanced
techniques applied in both medical and clinical settings. Cell therapies aim to treat and repair
the injury sites and replace the loss of tissues by stimulating the repair and regeneration process. In
order to enable the use of stem cells in regenerative therapies, further characterisation of cell behaviour,
in terms of their proliferation and differentiation capacity, mainly during the quiescent and inductive
state is regarded as highly necessary. The central focus of regenerative medicine revolves around
the use of human cells, including adult stem cells and induced pluripotent stem cells for cell-based
therapy. The purpose of this review was to examine the existing body of literature on stem cell research
conducted on cellular angiogenesis and migration, to investigate the validity of different strategies and
variations of the cell type used. The information gathered within this review may then be shared with
fellow researchers to assist in future research work, engaging in stem cell homing for cell-based therapy
to enhance wound healing and tissue regeneration process.
Collapse
Affiliation(s)
- Nur S. Aziz
- Postgraduate Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Norhayati Yusop
- Basic Sciences and Oral Biology Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Azlina Ahmad
- Basic Sciences and Oral Biology Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
43
|
Luo F, Zhang Z, Barnett A, Bellinger TJ, Turcato F, Schmidt K, Luo Y. Cuprizone-induced demyelination under physiological and post-stroke condition leads to decreased neurogenesis response in adult mouse brain. Exp Neurol 2020; 326:113168. [PMID: 31904386 DOI: 10.1016/j.expneurol.2019.113168] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 10/25/2022]
Abstract
Due to the limitation in treatment window of the rtPA (recombinant tissue plasminogen activator), the development of delayed treatment for stroke is needed. We previously reported that there is a difference in neurogenesis and neuroblast migration patterns in different mouse stroke models (proximal and distal middle cerebral artery occlusion models, pMCAo or dMCAo). Specifically, compared to robust neurogenesis and substantial migration of newly born neuroblasts in pMCAo model, dMCAo only illicit limited neurogenesis and migration of neuroblasts towards ischemic area. One potential reason for this difference is the relative location of ischemic area to white matter and the neurogenic niche (subventricular zone, SVZ). Specifically, white matter could serve as a physical barrier or inhibitory factor to neurogenesis and migration in the dMCAo model. Given that a major difference in human and rodent brains is the content of white matter in the brain, in this study, we further characterize these two models and test the important hypothesis that white matter is an important contributing inhibitory factor for the limited neurogenesis in the dMCAo model. We utilized a genetically inducible NSC-specific reporter mouse line (nestin-CreERT2-R26R-YFP) to label and track NSC proliferation, survival and differentiation in ischemic brain. To test whether myelin is inhibitory to neurogenesis in dMCAo model, we demyelinated mouse brains using cuprizone treatment after stroke and examined whether there is enhanced neurogenesis or migration of neuroblasts cells in stroke mice treated with cuprizone. Our data suggests that demyelination of the brain does not result in enhanced neurogenesis or migration of neuroblasts, supporting that myelin is not a major inhibitory factor for stroke-induced neurogenesis. In addition, our results suggest that in non-stroke mice, demyelination causes decreased neurogenesis in adult brain, indicating a potential positive role of myelin in maintenance of adult neural stem cell niche.
Collapse
Affiliation(s)
- Fucheng Luo
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, USA
| | - Zhen Zhang
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, USA
| | - Austin Barnett
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Tania J Bellinger
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, USA
| | - Flavia Turcato
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, USA
| | - Kelly Schmidt
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, USA
| | - Yu Luo
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, USA.
| |
Collapse
|
44
|
Decellularized brain matrix enhances macrophage polarization and functional improvements in rat spinal cord injury. Acta Biomater 2020; 101:357-371. [PMID: 31711898 DOI: 10.1016/j.actbio.2019.11.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/03/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is a devastating lesion lacking effective treatment options currently available in clinics. The inflammatory process exacerbates the extent of the lesion through a secondary injury mechanism, where proinflammatory classically activated macrophages (M1) are prevalent at the lesion site. However, the polarized alternatively activated anti-inflammatory macrophages (M2) are known to play an important role in wound healing and regeneration following SCI. Herein, we introduce porcine brain decellularized extracellular matrix (dECM) to modulate the macrophages in the injured spinal cord. The hydrogels with collagen and dECM at various dECM concentrations (1, 5, and 8 mg/ml) were used to cultivate primary macrophages and neurons. The dECM hydrogels were shown to promote the polarization of macrophages toward M2 phase and the neurite outgrowth of cortical and hippocampal neurons. When the dECM hydrogels were applied to rat SCI models, the proportion of M1 and M2 macrophages in the injured spinal cord was substantially altered. When received dECM concetration of 5 mg/ml, the expression of molecules associated with M2 (CD206, arginase1, and IL-10) was significantly increased. Consistently, the population of total macrophages and cavity area were substantially reduced in the dECM-treated groups. As a result, the locomotor functions of injured spinal cord, as assessed by BBB and ladder scoring, were significantly improved. Collectively, the porcine brain dECM with optimal concentration promotes functional recovery in SCI models through the activation of M2 macrophages, suggesting the promising use of the engineered hydrogels in the treatment of acute SCI. STATEMENT OF SIGNIFICANCE: Spinal cord injury (SCI) is a devastating lesion, lacking effective treatment options currently available in clinics. Here we delineated that the treatment of injured spinal cord with porcine brain decellularized matrix-based hydrogels for the first time, and could modulate the macrophage polarization and the ultimate functional recovery. When appropriate formulations were applied to a contused spinal cord model in rats, the decellularized matrix hydrogels shifted the macrophages to polarize to pro-regenerative M2 phenotype, decreased the size of lesion cavity, and finally promoted the locomotor functions until 8 weeks following the injury. We consider this work can significantly augment the matrix(biomaterial)-based therapeutic options, as an alternative to drug or cell-free approaches, for the treatment of acute injury of spinal cord.
Collapse
|
45
|
Abstract
Increased microvessel density in the peri-infarct region has been reported and has been correlated with longer survival times in ischemic stroke patients and has improved outcomes in ischemic animal models. This raises the possibility that enhancement of angiogenesis is one of the strategies to facilitate functional recovery after ischemic stroke. Blood vessels and neuronal cells communicate with each other using various mediators and contribute to the pathophysiology of cerebral ischemia as a unit. In this mini-review, we discuss how angiogenesis might couple with axonal outgrowth/neurogenesis and work for functional recovery after cerebral ischemia. Angiogenesis occurs within 4 to 7 days after cerebral ischemia in the border of the ischemic core and periphery. Post-ischemic angiogenesis may contribute to neuronal remodeling in at least two ways and is thought to contribute to functional recovery. First, new blood vessels that are formed after ischemia are thought to have a role in the guidance of sprouting axons by vascular endothelial growth factor and laminin/β1-integrin signaling. Second, blood vessels are thought to enhance neurogenesis in three stages: 1) Blood vessels enhance proliferation of neural stem/progenitor cells by expression of several extracellular signals, 2) microvessels support the migration of neural stem/progenitor cells toward the peri-infarct region by supplying oxygen, nutrients, and soluble factors as well as serving as a scaffold for migration, and 3) oxygenation induced by angiogenesis in the ischemic core is thought to facilitate the differentiation of migrated neural stem/progenitor cells into mature neurons. Thus, the regions of angiogenesis and surrounding tissue may be coupled, representing novel treatment targets.
Collapse
Affiliation(s)
- Masahiro Hatakeyama
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Itaru Ninomiya
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
46
|
Song D, Zhang X, Chen J, Liu X, Xue J, Zhang L, Lan X. Wnt canonical pathway activator TWS119 drives microglial anti-inflammatory activation and facilitates neurological recovery following experimental stroke. J Neuroinflammation 2019; 16:256. [PMID: 31810470 PMCID: PMC6896312 DOI: 10.1186/s12974-019-1660-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/26/2019] [Indexed: 11/29/2022] Open
Abstract
Background Ischemic stroke is a leading cause of disability worldwide and characteristically accompanied by downregulation of the Wnt/β-catenin signaling. Activation of Wnt/β-catenin signaling emerges to attenuate neuroinflammation after ischemic stroke; however, its effect on modulating microglial polarization is largely unknown. Here, we explored whether Wnt/β-catenin pathway activator TWS119 facilitated long-term neurological recovery via modulating microglia polarization after experimental stroke. Methods Ischemic stroke mice model was induced by permanent distal middle cerebral artery occlusion plus 1 h hypoxia. TWS119 was administrated from day 1 to 14 after stroke. Neurological deficits were monitored up to 21 days after stroke. Angiogenesis, neural plasticity, microglial polarization, and microglia-associated inflammatory cytokines were detected in the peri-infarct cortex at days 14 and 21 after stroke. Primary microglia and mouse brain microvascular endothelial cell lines were employed to explore the underlying mechanism in vitro. Results TWS119 mitigated neurological deficits at days 14 and 21 after experimental stroke, paralleled by acceleration on angiogenesis and neural plasticity in the peri-infarct cortex. Mechanistically, cerebral ischemia induced production of microglia-associated proinflammatory cytokines and priming of activated microglia toward pro-inflammatory polarization, whereas TWS119 ameliorated microglia-mediated neuroinflammatory status following ischemic stroke and promoted angiogenesis by modulating microglia to anti-inflammatory phenotype. The beneficial efficacy of TWS119 in microglial polarization was largely reversed by selective Wnt/β-catenin pathway blockade in vitro, suggesting that TWS119-enabled pro-inflammatory to anti-inflammatory phenotype switch of microglia was possibly mediated by Wnt/β-catenin signaling. Conclusions Wnt/β-catenin pathway activator TWS119 ameliorated neuroinflammatory microenvironment following chronic cerebral ischemia via modulating microglia towards anti-inflammatory phenotype, and facilitates neurological recovery in an anti-inflammatory phenotype polarization-dependent manner. Activation of Wnt/β-catenin pathway following ischemic stroke might be a potential restorative strategy targeting microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Degang Song
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Department of Neurology, First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China. .,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.
| | - Junmin Chen
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaoxia Liu
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Jing Xue
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xifa Lan
- Department of Neurology, First Hospital of Qinhuangdao, Qinhuangdao, 066000, Hebei, China
| |
Collapse
|
47
|
Gonzalez-Reyes LE, Chiang CC, Zhang M, Johnson J, Arrillaga-Tamez M, Couturier NH, Reddy N, Starikov L, Capadona JR, Kottmann AH, Durand DM. Sonic Hedgehog is expressed by hilar mossy cells and regulates cellular survival and neurogenesis in the adult hippocampus. Sci Rep 2019; 9:17402. [PMID: 31758070 PMCID: PMC6874678 DOI: 10.1038/s41598-019-53192-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Sonic hedgehog (Shh) is a multifunctional signaling protein governing pattern formation, proliferation and cell survival during embryogenesis. In the adult brain, Shh has neurotrophic function and is implicated in hippocampal neurogenesis but the cellular source of Shh in the hippocampus remains ill defined. Here, we utilize a gene expression tracer allele of Shh (Shh-nlacZ) which allowed the identification of a subpopulation of hilar neurons known as mossy cells (MCs) as a prominent and dynamic source of Shh within the dentate gyrus. AAV-Cre mediated ablation of Shh in the adult dentate gyrus led to a marked degeneration of MCs. Conversely, chemical stimulation of hippocampal neurons using the epileptogenic agent kainic acid (KA) increased the number of Shh+ MCs indicating that the expression of Shh by MCs confers a survival advantage during the response to excitotoxic insults. In addition, ablation of Shh in the adult dentate gyrus led to increased neural precursor cell proliferation and their migration into the subgranular cell layer demonstrating that MCs-generated Shh is a key modulator of hippocampal neurogenesis.
Collapse
Affiliation(s)
- Luis E Gonzalez-Reyes
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA.
- Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, Rehab. R&D, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH, 44106, USA.
| | - Chia-Chu Chiang
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Mingming Zhang
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Joshua Johnson
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Manuel Arrillaga-Tamez
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Nicholas H Couturier
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Neha Reddy
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Lev Starikov
- Department of Molecular, Cellular and Biomedical Sciences, CUNY School of Medicine at City College of New York and Graduate Center, City University of New York, New York, NY, 10031, USA
| | - Jeffrey R Capadona
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
- Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, Rehab. R&D, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH, 44106, USA
| | - Andreas H Kottmann
- Department of Molecular, Cellular and Biomedical Sciences, CUNY School of Medicine at City College of New York and Graduate Center, City University of New York, New York, NY, 10031, USA
| | - Dominique M Durand
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
48
|
Giarretta I, Gaetani E, Bigossi M, Tondi P, Asahara T, Pola R. The Hedgehog Signaling Pathway in Ischemic Tissues. Int J Mol Sci 2019; 20:ijms20215270. [PMID: 31652910 PMCID: PMC6862352 DOI: 10.3390/ijms20215270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022] Open
Abstract
Hedgehog (Hh) proteins are prototypical morphogens known to regulate epithelial/mesenchymal interactions during embryonic development. In addition to its pivotal role in embryogenesis, the Hh signaling pathway may be recapitulated in post-natal life in a number of physiological and pathological conditions, including ischemia. This review highlights the involvement of Hh signaling in ischemic tissue regeneration and angiogenesis, with particular attention to the heart, the brain, and the skeletal muscle. Updated information on the potential role of the Hh pathway as a therapeutic target in the ischemic condition is also presented.
Collapse
Affiliation(s)
- Igor Giarretta
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Eleonora Gaetani
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Margherita Bigossi
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Paolo Tondi
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.
| | - Roberto Pola
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
49
|
Qin S, Sun D, Zhang C, Tang Y, Zhou F, Zheng K, Tang R, Zheng Y. Downregulation of sonic hedgehog signaling in the hippocampus leads to neuronal apoptosis in high-fat diet-fed mice. Behav Brain Res 2019; 367:91-100. [DOI: 10.1016/j.bbr.2019.03.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/28/2019] [Accepted: 03/30/2019] [Indexed: 12/15/2022]
|
50
|
Zhong J, Lan C, Zhang C, Yang Y, Chen WX, Zhang KY, Zhao HL, Fang XY, Li HH, Tan L, Wang P, Ge HF, Hu R, Feng H. Chondroitin sulfate proteoglycan represses neural stem/progenitor cells migration via PTPσ/α-actinin4 signaling pathway. J Cell Biochem 2019; 120:11008-11021. [PMID: 30688376 DOI: 10.1002/jcb.28379] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
Neural stem/progenitor cells (NSPCs) are a promising candidate for the cell-replacement therapy after central nervous system (CNS) injury. However, the short of sufficient NSPCs migration and integration into the lesions is an essential challenge for cell-based therapy after CNS injury due to the disturbance of local environmental homeostasis. Chondroitin sulfate proteoglycan (CSPG) is obviously accumulated at the lesions and destroyed local homeostasis after CNS injury. The previous study has demonstrated that the CSPG is a dominating ingredient inhibiting axonal regrowth of newly born neurons after CNS injury. NSPCs, a strain of special neural subtypes, hold the capacity of leading processes formation to regulate NSPCs migration, which has the same mechanism as axonal regrowth. Hence, it is worth investigating the effect of CSPG on NSPCs migration and its underlying mechanism. Here, different concentration of CSPG was used to evaluate its effect on NSPCs migration. The results showed that the CSPG suppressed NSPCs migration in a dose-dependent manner from 10 to 80 µg/mL with phase-contrast microscopy after 24 hours. Meanwhile, transwell assays were performed to certify the above results. Our data indicated that the 40 µg/mL CSPG obviously suppressed NSPCs migration via decreasing filopodia formation using immunofluorescence staining. Furthermore, data indicated that the 40 µg/mL CSPG upregulated protein tyrosine phosphatase receptor σ (PTPσ) expression and decreased α-actinin4 (ACTN4) expression through immunofluorescence, reverse transcription polymerase chain reaction, and Western blot assays. While the inhibitory effect was attenuated using PTPσ-specific small interfering RNA. In addition, data demonstrated that the 40 µg/mL CSPG facilitated NSPCs differentiation into glial fibrillary acidic protein-positive cells and inhibited NSPCs directing into MAP2- and MBP-positive cells. Collectively, these data demonstrated that the CSPG suppressed NSPCs migration through PTPσ/ACTN4 signaling pathway. Meanwhile, CSPG facilitated NSPCs differentiation into astrocytes and inhibited NSPCs directing into neurons and oligodendrocytes.
Collapse
Affiliation(s)
- Jun Zhong
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuan Lan
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chao Zhang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei-Xiang Chen
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Kai-Yuan Zhang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Heng-Li Zhao
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuan-Yu Fang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huan-Huan Li
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liang Tan
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pan Wang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hong-Fei Ge
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rong Hu
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|