1
|
Bhardwaj K, Jha A, Roy A, Kumar H. The crucial role of VPS35 and SHH in Parkinson's disease: Understanding the mechanisms behind the neurodegenerative disorder. Brain Res 2024; 1845:149204. [PMID: 39197569 DOI: 10.1016/j.brainres.2024.149204] [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: 04/28/2024] [Revised: 07/10/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Parkinson's disease (PD) is indeed a complex neurodegenerative disorder recognized by the progressive depletion of dopaminergic neurons in the brain, particularly in the substantia nigra region, leading to motor impairments and other symptoms. But at the molecular level, the study about PD still lacks. As the number of cases worldwide continues to increase, it is critical to focus on the cellular and molecular mechanisms of the disease's presentation and neurodegeneration to develop novel therapeutic approaches. At the molecular level, the complexity is more due to the involvement of vacuolar protein sorting 35 (VPS35) and sonic hedgehog (SHH) signaling in PD (directly or indirectly), leading to one of the most prominent hallmarks of the disease, which is an accumulation of α-synuclein. This elevated pathogenesis may result from impaired autophagy due to mutation in the case of VPS35 and impairment in SHH signaling at the molecular level. The traditional understanding of PD is marked by the disruption of dopaminergic neurons and dopaminergic signaling, which exacerbates symptoms of motor function deficits. However, the changes at the molecular level that are being disregarded also impact the overall health of the dopaminergic system. Gaining insight into these two unique mechanisms is essential to determine whether they give neuroprotection or have no effect on the health of neurons. Hence, here we tried to simplify the understanding of the role of VPS35 and SHH signaling to comprehend it in one direction.
Collapse
Affiliation(s)
- Kritika Bhardwaj
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Akanksha Jha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
2
|
Prajapati A, Mehan S, Khan Z, Chhabra S, Das Gupta G. Purmorphamine, a Smo-Shh/Gli Activator, Promotes Sonic Hedgehog-Mediated Neurogenesis and Restores Behavioural and Neurochemical Deficits in Experimental Model of Multiple Sclerosis. Neurochem Res 2024; 49:1556-1576. [PMID: 38160216 DOI: 10.1007/s11064-023-04082-9] [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: 04/18/2023] [Revised: 09/27/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
Multiple sclerosis (MS) is a pathological condition characterized by the demyelination of nerve fibers, primarily attributed to the destruction of oligodendrocytes and subsequent motor neuron impairment. Ethidium bromide (EB) is a neurotoxic compound that induces neuronal degeneration, resulting in demyelination and symptoms resembling those observed in experimental animal models of multiple sclerosis (MS). The neurotoxic effects induced by EB in multiple sclerosis (MS) are distinguished by the death of oligodendrocytes, degradation of myelin basic protein (MBP), and deterioration of axons. Neurological complications related to MS have been linked to alterations in the signaling pathway known as smo-shh. Purmorphine (PUR) is a semi-synthetic compound that exhibits potent Smo-shh agonistic activity. It possesses various pharmacological properties, including antioxidant, anti-inflammatory, anti-apoptotic, and neuromodulatory effects. Hence, the current investigation was conducted to assess the neuroprotective efficacy of PUR (at doses of 5 and 10 mg/kg, administered intraperitoneally) both individually and in conjunction with Fingolimod (FING) (at a dose of 0.5 mg/kg, administered intraperitoneally) in the experimental model of MS induced by EB. The administration of EB was conducted via the intracerebropeduncle route (ICP) over a period of seven days in the brain of rats. The Wistar rats were allocated into six groups using randomization, each consisting of eight rats (n = 8 per group). The experimental groups in this study were categorized as follows: (I) Sham Control, (II) Vehicle Control, (III) PUR per se, (IV) EB, (V) EB + PUR5, (VI) EB + PUR10, (VII) EB + FING 0.5, and (VIII) EB + PUR10 + FING 0.5. On the final day of the experimental timeline, all animal subjects were euthanized, and subsequent neurochemical estimations were conducted on cerebrospinal fluid, blood plasma, and brain tissue samples. In addition, we conducted neurofilament (NFL) analysis and histopathological examination. We utilized the luxol myelin stain to understand better the degeneration associated with MS and its associated neurological complications. The findings of our study indicate that the activation of SMO-Shh by PUR has a mitigating effect on neurobehavioral impairments induced by EB, as well as a restorative effect on cellular and neurotransmitter abnormalities in an experimental model of MS.
Collapse
Affiliation(s)
- Aradhana Prajapati
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India.
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), NAAC Accredited "A" Grade College, GT Road, Ghal-Kalan, Moga, Punjab, 142001, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Swesha Chhabra
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
- IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| |
Collapse
|
3
|
Guo S, Lei Q, Yang Q, Chen R. IGFBP5 Promotes Neuronal Apoptosis in a 6-OHDA-Toxicant Model of Parkinson's Disease by Inhibiting the Sonic Hedgehog Signaling Pathway. Med Princ Pract 2024; 33:269-280. [PMID: 38565090 PMCID: PMC11175608 DOI: 10.1159/000538467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Parkinson's disease (PD) is the most common neurodegenerative disease worldwide. Studies have shown that insulin-like growth factor-binding protein 5 (IGFBP5) may contribute to methamphetamine-induced neurotoxicity and neuronal apoptosis in PC-12 cells and rat striatum. Here, we studied the expression and role of IGFBP5 in the 6-OHDA-toxicant model of PD. METHODS PC-12 and SH-SY5Y cells were exposed to 50 μm 6-OHDA for 24 h. qRT-PCR, western blotting, CCK-8 assay, EdU staining, annexin V staining, and immunofluorescence were performed to study the effects of IGFBP5-specific siRNAs. The effects of IGFBP5 on a rat 6-OHDA model of PD were confirmed by performing behavioral tests, tyrosine hydroxylase (TH) immunofluorescence staining, and western blotting. RESULTS In the GSE7621 dataset, IGFBP5 was highly expressed in the substantia nigra tissues of PD patients compared to healthy controls. In PC-12 and SH-SY5Y cells, IGFBP5 was upregulated following 6-OHDA exposure in a dose-dependent manner. Silencing of IGFBP5 promoted PC-12 and SH-SY5Y proliferation and inhibited apoptosis under 6-OHDA stimulation. Silencing of IGFBP5 relieved 6-OHDA-induced TH-positive neuron loss. Hedgehog signaling pathway was predicted as a downstream signaling pathway of IGFBP5. Negative regulation between IGFBP5 and sonic hedgehog (SHH) signaling pathway was confirmed in vitro. The effects of IGFBP5 silencing on SH-SY5Y cells were partially reversed using cyclopamine, a direct inhibitor of the SHH signaling pathway. In addition, silencing of IGFBP5 attenuated motor deficits and neuronal damage in 6-OHDA-induced PD rats. CONCLUSION Elevated IGFBP5 expression may be involved in 6-OHDA-induced neurotoxicity through regulation of the SHH signaling pathway.
Collapse
Affiliation(s)
- Shenglong Guo
- Shaanxi Provincial People's Hospital, Department II of Neurology, Xi'an, China
| | - Qi Lei
- Shaanxi Provincial People's Hospital, Department II of Neurology, Xi'an, China
| | - Qian Yang
- Shaanxi Provincial People's Hospital, Department II of Neurology, Xi'an, China
| | - Ruili Chen
- Shaanxi Provincial People's Hospital, Department II of Neurology, Xi'an, China
| |
Collapse
|
4
|
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: 13] [Impact Index Per Article: 6.5] [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
|
5
|
Prakash N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front Mol Neurosci 2022; 15:1071731. [PMID: 36618829 PMCID: PMC9815185 DOI: 10.3389/fnmol.2022.1071731] [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: 10/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson's Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Collapse
|
6
|
Cai Y, Zhang MM, Wang M, Jiang ZH, Tan ZG. Bone Marrow-Derived Mesenchymal Stem Cell-Derived Exosomes Containing Gli1 Alleviate Microglial Activation and Neuronal Apoptosis In Vitro and in a Mouse Parkinson Disease Model by Direct Inhibition of Sp1 Signaling. J Neuropathol Exp Neurol 2022; 81:522-534. [PMID: 35609560 DOI: 10.1093/jnen/nlac037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
This study investigated possible therapeutic effect mechanisms of exosomes from bone marrow-derived mesenchymal stem cells (BMSC) in neuronal and microglial cells and in a Parkinson disease (PD) model. Neuronal SH-SY5Y cells and microglial HMC3 cells were subjected to 1-methyl-4-phenylpyridinium (MPP+) or LPS, respectively. The mRNA and protein expression was assessed using qRT-PCR, Western blotting, and enzyme-linked immunosorbent assay. Cell viability and apoptosis of SH-SY5Y cells were examined using the MTT assay and flow cytometry. Chromatin immunoprecipitation assays were performed to assess the binding relationship between glioma-associated oncogene homolog 1 (Gli1) and the Sp1 transcription factor promoter. BMSC-derived exosomes promoted cell proliferation and inhibited apoptosis in MPP+-treated SH-SY5Y cells and suppressed inflammatory markers in LPS-treated HMC3 cells. Sp1 knockdown decreased SH-SY5Y cell damage and HMC3 immune activation. Gli1 carried by BMSC exosomes directly bound with Sp1 to inhibit Sp1-mediated LRRK2 activation whereas exosomes secreted by Gli1-knockdown in BMSC did not. In a PD mouse model induced with MPTP, BMSC exosomes decreased neuron loss injury and the inflammatory response by inhibiting Sp1 signaling. Thus, BMSC-derived exosomal Gli1 alleviates inflammatory damage and neuronal apoptosis by inhibiting Sp1 in vitro and in vivo. These findings provide the basis for the potential clinical use of BMSC-derived exosomes in PD.
Collapse
Affiliation(s)
- Yang Cai
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Ming-Ming Zhang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Ming Wang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Zhuo-Hang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| | - Zhi-Gang Tan
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011, P.R. China
| |
Collapse
|
7
|
Zhang J, Wang X, Bernardi RE, Ju J, Wei S, Gong Z. Activation of AMPA Receptors in the Lateral Habenula Produces Anxiolytic Effects in a Rat Model of Parkinson’s Disease. Front Pharmacol 2022; 13:821975. [PMID: 35145415 PMCID: PMC8822149 DOI: 10.3389/fphar.2022.821975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/07/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Parkinson’s disease (PD) is commonly accompanied with anxiety disorder, however, the mechanisms underlying PD-mediated anxiety remain elusive. The lateral habenula (LHb) is a critical brain region that influences the activity of the monoaminergic system in the midbrain and consequently modulates anxiety. Most neurons in the LHb express AMPA receptors (AMPARs). The PD model for the pharmacological intervention of AMPA receptors was established by the unilateral lesion of the substantia nigra pars compacta (SNc) with 6-hydroxydopamine (6-OHDA). Methods: The AMPAR agonist (S)-AMPA and antagonist NBQX were microinjected into the LHb, respectively, to examine whether anxiety-like behaviors were altered in sham-operated and SNc-lesion rats, measured with the paradigms of the open-field test (OPT) and elevated plus maze (EPM). Furthermore, dopamine (DA) and 5-hydroxytryptamine (5-HT) levels in the basolateral amygdala (BLA) were measured using in vivo microdialysis immediately following the injections of (S)-AMPA and NBQX into the LHb. Results: Activation of LHb AMPA receptors by (S)-AMPA produced anxiolytic-like behaviors and enhanced the extracellular DA and 5-HT in the BLA. Conversely, NBQX induced anxiety-like effects and suppressed the extracellular DA and 5-HT in the BLA. In addition, the minimal doses inducing the effects in the SNc-lesion rats were lower than those in sham-operated rats. Conclusion: These findings suggest that the effects of AMPA receptors in the LHb on anxiety-like behaviors likely involve the extracellular levels of DA and 5-HT in the BLA. The present results may improve our understanding of the neuropathology and/or treatment of PD.
Collapse
Affiliation(s)
- Jin Zhang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Shenzhen Graduate School, Peking University, Shenzhen, China
| | - Xiaobing Wang
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
| | - Rick E. Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg, Germany
| | - Jun Ju
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Shoupeng Wei
- Tomas Lindahl Nobel Laureate Laboratory, Precision Medicine Research Centre, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Shoupeng Wei, ; Zhiting Gong,
| | - Zhiting Gong
- Department of Anatomy, College of Preclinical Medicine, Dali University, Dali, China
- *Correspondence: Shoupeng Wei, ; Zhiting Gong,
| |
Collapse
|
8
|
Ma R, Kutchy NA, Chen L, Meigs DD, Hu G. Primary cilia and ciliary signaling pathways in aging and age-related brain disorders. Neurobiol Dis 2022; 163:105607. [PMID: 34979259 PMCID: PMC9280856 DOI: 10.1016/j.nbd.2021.105607] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Brain disorders are characterized by the progressive loss of structure and function of the brain as a consequence of progressive degeneration and/or death of nerve cells. Aging is a major risk factor for brain disorders such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and stroke. Various cellular and molecular events have been shown to play a role in the progress of neurodegenerative diseases. Emerging studies suggest that primary cilia could be a key regulator in brain diseases. The primary cilium is a singular cellular organelle expressed on the surface of many cell types, such as astrocytes and neurons in the mature brain. Primary cilia detect extracellular cues, such as Sonic Hedgehog (SHH) protein, and transduce these signals into cells to regulate various signaling pathways. Abnormalities in ciliary length and frequency (ratio of ciliated cells) have been implicated in various human diseases, including brain disorders. This review summarizes current findings and thoughts on the role of primary cilia and ciliary signaling pathways in aging and age-related brain disorders.
Collapse
Affiliation(s)
- Rong Ma
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Naseer A Kutchy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Anatomy, Physiology and Pharmacology, School of Veterinary Medicine, St. George's University, Grenada
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, Guangdong 515063, China; Key Laboratory of Intelligent Manufacturing Technology, Ministry of Education, Shantou University, Shantou, Guangdong 515063, China
| | - Douglas D Meigs
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
9
|
Integrated microarray analysis provided a new insight of the pathogenesis of Parkinson’s disease. Neurosci Lett 2018; 662:51-58. [DOI: 10.1016/j.neulet.2017.09.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 08/25/2017] [Accepted: 09/25/2017] [Indexed: 12/14/2022]
|
10
|
Ugbode CI, Smith I, Whalley BJ, Hirst WD, Rattray M. Sonic hedgehog signalling mediates astrocyte crosstalk with neurons to confer neuroprotection. J Neurochem 2017; 142:429-443. [PMID: 28485896 PMCID: PMC5575469 DOI: 10.1111/jnc.14064] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/18/2017] [Accepted: 05/03/2017] [Indexed: 01/10/2023]
Abstract
Sonic hedgehog (SHH) is a glycoprotein associated with development that is also expressed in the adult CNS and released after brain injury. Since the SHH receptors patched homolog‐1 and Smoothened are highly expressed on astrocytes, we hypothesized that SHH regulates astrocyte function. Primary mouse cortical astrocytes derived from embryonic Swiss mouse cortices, were treated with two chemically distinct agonists of the SHH pathway, which caused astrocytes to elongate and proliferate. These changes are accompanied by decreases in the major astrocyte glutamate transporter‐1 and the astrocyte intermediate filament protein glial fibrillary acidic protein. Multisite electrophysiological recordings revealed that the SHH agonist, smoothened agonist suppressed neuronal firing in astrocyte‐neuron co‐cultures and this was abolished by the astrocyte metabolic inhibitor ethylfluoroacetate, revealing that SHH stimulation of metabolically active astrocytes influences neuronal firing. Using three‐dimensional co‐culture, MAP2 western blotting and immunohistochemistry, we show that SHH‐stimulated astrocytes protect neurons from kainate‐induced cell death. Altogether the results show that SHH regulation of astrocyte function represents an endogenous neuroprotective mechanism. ![]()
Collapse
Affiliation(s)
- Christopher I Ugbode
- School of Pharmacy, University of Bradford, Bradford, UK.,School of Chemistry, Food & Pharmacy, University of Reading, Reading, UK.,Department of Biology, University of York, Heslington, UK
| | - Imogen Smith
- School of Chemistry, Food & Pharmacy, University of Reading, Reading, UK.,Portsmouth Brain Tumour Research Centre, University of Portsmouth, Portsmouth, UK
| | - Benjamin J Whalley
- School of Chemistry, Food & Pharmacy, University of Reading, Reading, UK
| | - Warren D Hirst
- Neurodegeneration and Neurologic Diseases, Pfizer Neuroscience Research Unit, Cambridge, Massachusetts, USA
| | - Marcus Rattray
- School of Pharmacy, University of Bradford, Bradford, UK
| |
Collapse
|
11
|
Purmorphamine as a Shh Signaling Activator Small Molecule Promotes Motor Neuron Differentiation of Mesenchymal Stem Cells Cultured on Nanofibrous PCL Scaffold. Mol Neurobiol 2016; 54:5668-5675. [DOI: 10.1007/s12035-016-0090-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/30/2016] [Indexed: 12/24/2022]
|
12
|
Towards a Better Treatment Option for Parkinson’s Disease: A Review of Adult Neurogenesis. Neurochem Res 2016; 41:3161-3170. [DOI: 10.1007/s11064-016-2053-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 02/04/2023]
|
13
|
Abstract
Neuroscience research has been revolutionized by the use of recombinant viral vector technology from the basic, preclinical and clinical levels. Currently, multiple recombinant viral vector types are employed with each having its strengths and weaknesses depending on the proposed application. Helper-dependent adenoviral vectors (HdAd) are emerging as ideal viral vectors that solve a major need in the neuroscience field: (1) expression of transgenes that are too large to be packaged by other viral vectors and (2) rapid onset of transgene expression in the absence of cytotoxicity. Here, we describe the methods for large-scale production of HdAd viral vectors for in vivo use with neurospecific transgene expression.
Collapse
|
14
|
Singh VK, Kalsan M, Kumar N, Saini A, Chandra R. Induced pluripotent stem cells: applications in regenerative medicine, disease modeling, and drug discovery. Front Cell Dev Biol 2015; 3:2. [PMID: 25699255 PMCID: PMC4313779 DOI: 10.3389/fcell.2015.00002] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/06/2015] [Indexed: 12/12/2022] Open
Abstract
Recent progresses in the field of Induced Pluripotent Stem Cells (iPSCs) have opened up many gateways for the research in therapeutics. iPSCs are the cells which are reprogrammed from somatic cells using different transcription factors. iPSCs possess unique properties of self renewal and differentiation to many types of cell lineage. Hence could replace the use of embryonic stem cells (ESC), and may overcome the various ethical issues regarding the use of embryos in research and clinics. Overwhelming responses prompted worldwide by a large number of researchers about the use of iPSCs evoked a large number of peple to establish more authentic methods for iPSC generation. This would require understanding the underlying mechanism in a detailed manner. There have been a large number of reports showing potential role of different molecules as putative regulators of iPSC generating methods. The molecular mechanisms that play role in reprogramming to generate iPSCs from different types of somatic cell sources involves a plethora of molecules including miRNAs, DNA modifying agents (viz. DNA methyl transferases), NANOG, etc. While promising a number of important roles in various clinical/research studies, iPSCs could also be of great use in studying molecular mechanism of many diseases. There are various diseases that have been modeled by uing iPSCs for better understanding of their etiology which maybe further utilized for developing putative treatments for these diseases. In addition, iPSCs are used for the production of patient-specific cells which can be transplanted to the site of injury or the site of tissue degeneration due to various disease conditions. The use of iPSCs may eliminate the chances of immune rejection as patient specific cells may be used for transplantation in various engraftment processes. Moreover, iPSC technology has been employed in various diseases for disease modeling and gene therapy. The technique offers benefits over other similar techniques such as animal models. Many toxic compounds (different chemical compounds, pharmaceutical drugs, other hazardous chemicals, or environmental conditions) which are encountered by humans and newly designed drugs may be evaluated for toxicity and effects by using iPSCs. Thus, the applications of iPSCs in regenerative medicine, disease modeling, and drug discovery are enormous and should be explored in a more comprehensive manner.
Collapse
Affiliation(s)
- Vimal K Singh
- INSPIRE Faculty, Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Manisha Kalsan
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Neeraj Kumar
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Ramesh Chandra
- B. R. Ambedkar Centre for Biomedical Research, University of Delhi Delhi, India
| |
Collapse
|
15
|
Petrova R, Joyner AL. Roles for Hedgehog signaling in adult organ homeostasis and repair. Development 2014; 141:3445-57. [PMID: 25183867 DOI: 10.1242/dev.083691] [Citation(s) in RCA: 290] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The hedgehog (HH) pathway is well known for its mitogenic and morphogenic functions during development, and HH signaling continues in discrete populations of cells within many adult mammalian tissues. Growing evidence indicates that HH regulates diverse quiescent stem cell populations, but the exact roles that HH signaling plays in adult organ homeostasis and regeneration remain poorly understood. Here, we review recently identified functions of HH in modulating the behavior of tissue-specific adult stem and progenitor cells during homeostasis, regeneration and disease. We conclude that HH signaling is a key factor in the regulation of adult tissue homeostasis and repair, acting via multiple different routes to regulate distinct cellular outcomes, including maintenance of plasticity, in a context-dependent manner.
Collapse
Affiliation(s)
- Ralitsa Petrova
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA BCMB Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Alexandra L Joyner
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Avenue, New York, NY 10065, USA BCMB Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| |
Collapse
|
16
|
Zhang Y, Dong W, Guo S, Zhao S, He S, Zhang L, Tang Y, Wang H. Lentivirus-mediated delivery of sonic hedgehog into the striatum stimulates neuroregeneration in a rat model of Parkinson disease. Neurol Sci 2014; 35:1931-40. [PMID: 25030123 DOI: 10.1007/s10072-014-1866-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/25/2014] [Indexed: 12/14/2022]
Abstract
Parkinson disease (PD) is a progressive neurodegenerative disorder in which the nigrostriatal pathway, consisting of dopaminergic neuronal projections from the substantia nigra to the striatum, degenerates. Viral transduction is currently the most promising in vivo strategy for delivery of therapeutic proteins into the brain for treatment of PD. Sonic hedgehog (Shh) is necessary for cell proliferation, differentiation and neuroprotection in the central nervous system. In this study, we investigated the effects of overexpressed N-terminal product of SHH (SHH-N) in a PD model rat. A lentiviral vector containing SHH-N was stereotactically injected into the striatum 24 h after a striatal 6-OHDA lesion. We found that overexpressed SHH-N attenuated behavioral deficits and reduced the loss of dopamine neurons in the substantia nigra and the loss of dopamine fibers in the striatum. In addition, fluoro-ruby-labeled nigrostriatal projections were also repaired. Together, our results demonstrate the feasibility and efficacy of using the strategy of lentivirus-mediated Shh-N delivery to delay nigrostriatal pathway degeneration. This strategy holds the potential for therapeutic application in the treatment of PD.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Gynecology and Obstetrics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Hadden MK. Hedgehog pathway agonism: therapeutic potential and small-molecule development. ChemMedChem 2013; 9:27-37. [PMID: 24203435 DOI: 10.1002/cmdc.201300358] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Indexed: 11/10/2022]
Abstract
The Hedgehog (Hh) pathway is a developmental signaling pathway that plays multiple roles during embryonic development and in adult tissues. Constitutive Hh signaling has been linked to the development and progression of several forms of cancer, and the application of small-molecule pathway inhibitors as anticancer chemotherapeutics is well studied and clearly defined. Activation of the Hh pathway as a therapeutic strategy for a variety of degenerative or ischemic disorders has also been proposed; however, the development of small-molecule Hh agonists has received less attention. The goal of this review is to highlight the recent evidence supporting the therapeutic potential of Hh pathway activators and to provide a comprehensive overview of small-molecule pathway agonists.
Collapse
Affiliation(s)
- M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, CT 06269 (USA).
| |
Collapse
|
18
|
Ma X, Turnbull P, Peterson R, Turnbull J. Trophic and proliferative effects of Shh on motor neurons in embryonic spinal cord culture from wildtype and G93A SOD1 mice. BMC Neurosci 2013; 14:119. [PMID: 24119209 PMCID: PMC3852546 DOI: 10.1186/1471-2202-14-119] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 09/18/2013] [Indexed: 12/01/2022] Open
Abstract
Background The developmental morphogen sonic hedgehog (Shh) may continue to play a trophic role in the support of terminally-differentiated motor neurons, of potential relevance to motor neuron disease. In addition, it may support the proliferation and differentiation of endogenous stem cells along motor neuronal lineages. As such, we have examined the trophic and proliferative effects of Shh supplementation or Shh antagonism in embryonic spinal cord cell cultures derived from wildtype or G93A SOD1 mice, a mouse model of amyotrophic lateral sclerosis. Results Shh supported survival, and stimulated growth of motor neurons, neurite outgrowth, and neurosphere formation in primary culture derived from both G93A SOD1 and WT mice. Shh increased the percentage of ciliated motor neurons, especially in G93A SOD1 culture. Shh-treated cultures showed increased neuronal proliferation compared to controls and especially cyclopamine treated cultures, from G93A SOD1 and WT mice. Moreover, Shh enhanced cell survival and differentiation of motor neuron precursors in WT culture. Conclusions Shh is neurotrophic to motor neurons and has mitogenic effects in WT and mSOD1 G93A culture in vitro.
Collapse
Affiliation(s)
- Xiaoxing Ma
- Department of Medicine, McMaster University, 1200 Main St West, Hamilton, ON L8N 3Z5, Canada.
| | | | | | | |
Collapse
|
19
|
Bashir S, Halepoto DM, Al-Ayadhi L. Serum level of desert hedgehog protein in autism spectrum disorder: preliminary results. Med Princ Pract 2013; 23:14-7. [PMID: 24009062 PMCID: PMC5586831 DOI: 10.1159/000354295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 07/11/2013] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To investigate the role of desert hedgehog (Dhh) in a neurodevelopmental disorder known as autism. SUBJECTS AND METHODS This study was conducted at the Autism Research and Treatment Center, King Khalid University Hospital, Riyadh, Kingdom of Saudi Arabia from October 2011 to May 2012. The serum levels of the Dhh protein in 57 patients recently diagnosed with autism and 37 age-matched healthy children were measured using ELISA. The Childhood Autism Rating Scale (CARS) was used for the assessment of autistic severity. RESULTS The mean serum level of Dhh in patients with autism (1.38 ± 0.50 ng/ml) was significantly lower (p = 0.0003) than that of normal controls (1.73 ± 0.37 ng/ml). There was no significant relationship between the serum level of Dhh and the CARS score (p = 0.28), age (p = 0.51) or gender (p = 0.76). CONCLUSIONS The Dhh serum level of patients with autism was lower than that of controls, probably indicating that the serum level of Dhh might be implicated in the physiology of autism. However, this finding should be treated with caution until further investigations are performed with larger populations.
Collapse
Affiliation(s)
- Shahid Bashir
- Division of Cognitive Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass., USA
- KSU-Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Dost Muhammad Halepoto
- KSU-Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- KSU-Autism Research and Treatment Center, Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Masjkur J, Rueger MA, Bornstein SR, McKay R, Androutsellis-Theotokis A. Neurovascular signals suggest a propagation mechanism for endogenous stem cell activation along blood vessels. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2013; 11:805-17. [PMID: 23131162 PMCID: PMC3580829 DOI: 10.2174/1871527311201070805] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/20/2012] [Accepted: 08/15/2012] [Indexed: 12/27/2022]
Abstract
Stem cell – based therapies for central nervous system disorders are intensely pursued. Such approaches can be divided into two categories: Transplantation-based, and those that aim to pharmacologically target the endogenous stem cell population in the tissue. Endogenous stem cell – based strategies avoid the problem of immune incompatibility between the host and the grafted cells. They also avoid the placement of a large amount of cells in confined areas, a manipulation which alters the characteristics of the neurovascular microenvironment. We show here that massive pharmacological activation (increase in cell numbers) of the endogenous neural stem cell population in the adult rodent brain maintains the cytoarchitecture of the neurovascular niche. Distances between adjacent stem cells (identified by expression of Hes3) are maintained above a minimum. Hes3+ cells maintain their physical association with blood vessels. These results also suggest a mechanism by which the activation signal from the lateral ventricle can be propagated to areas a long distance away from the lateral ventricles, through autocrine/paracrine actions between adjacent Hes3+ cells, along blood vessels. Finally, powerful effects of angiopoietin 2 on Hes3+ cells help explain the prevalence of proliferating endogenous neural stem cells close to the subventricular zone (an area of high angiopoietin 2 concentration) and the quiescent state of stem cells away from the ventricles and their tight physical association with blood vessels (which express high levels of angiopoietin 1, a cytokine that opposes angiopoietin 2 functions).
Collapse
Affiliation(s)
- Jimmy Masjkur
- Department of Medicine, University of Dresden, Dresden, Germany
| | | | | | | | | |
Collapse
|
21
|
Lin HY, Yeh WL, Huang BR, Lin C, Lai CH, Lin H, Lu DY. Desipramine protects neuronal cell death and induces heme oxygenase-1 expression in Mes23.5 dopaminergic neurons. PLoS One 2012; 7:e50138. [PMID: 23209658 PMCID: PMC3507930 DOI: 10.1371/journal.pone.0050138] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 10/17/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Desipramine is known principally as a tricyclic antidepressant drug used to promote recovery of depressed patients. It has also been used in a number of other psychiatric and medical conditions. The present study is the first to investigate the neuroprotective effect of desipramine. METHODOLOGY/PRINCIPAL FINDINGS Mes23.5 dopaminergic cells were used to examine neuroprotective effect of desipramine. Western blot, reverse transcription-PCR, MTT assay, siRNA transfection and electrophoretic mobility shift assay (EMSA) were carried out to assess the effects of desipramine. Desipramine induces endogenous anti-oxidative enzyme, heme oxygenase-1 (HO-1) protein and mRNA expression in concentration- and time-dependent manners. A different type of antidepressant SSRI (selective serotonin reuptake inhibitor), fluoxetine also shows similar effects of desipramine on HO-1 expression. Moreover, desipramine induces HO-1 expression through activation of ERK and JNK signaling pathways. Desipramine also increases NF-E2-related factor-2 (Nrf2) accumulation in the nucleus and enhances Nrf2-DNA binding activity. Moreover, desipramine-mediated increase of HO-1 expression is reduced by transfection with siRNA against Nrf2. On the other hand, pretreatment of desipramine protects neuronal cells against rotenone- and 6-hydroxydopamine (6-OHDA)-induced neuronal death. Furthermore, inhibition of HO-1 activity by a HO-1 pharmacological inhibitor, ZnPP IX, attenuates the neuroprotective effect of desipramine. Otherwise, activation of HO-1 activity by HO-1 activator and inducer protect 6-OHDA-induced neuronal death. CONCLUSIONS/SIGNIFICANCE These findings suggest that desipramine-increased HO-1 expression is mediated by Nrf2 activation through the ERK and JNK signaling pathways. Our results also suggest that desipramine provides a novel effect of neuroprotection, and neurodegenerative process might play an important role in depression disorder.
Collapse
Affiliation(s)
- Hsiao-Yun Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Lan Yeh
- Cancer Research Center, Department of Medical Research, Changhua Christian Hospital, Changhua, Taiwan
| | - Bor-Ren Huang
- Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Taichung Branch, Taichung, Taiwan
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- * E-mail: (HL); (D-YL)
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung, Taiwan
- * E-mail: (HL); (D-YL)
| |
Collapse
|
22
|
Fuchs J, Stettler O, Alvarez-Fischer D, Prochiantz A, Moya KL, Joshi RL. Engrailed signaling in axon guidance and neuron survival. Eur J Neurosci 2012; 35:1837-45. [PMID: 22708594 DOI: 10.1111/j.1460-9568.2012.08139.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several homeoproteins can function in a direct cell non-autonomous fashion to control various biological processes. In the developing nervous system, this mode of signaling has been well documented for Engrailed in the guidance of retinal ganglion cell axons and retino-tectal patterning. Engrailed is also a key factor for mesencephalic dopaminergic (mDA) neurons, not only during development but also in the adult. Haplodeficiency for Engrailed1 leads to progressive adult-onset loss of mDA neurons and several phenotypic alterations reminiscent of Parkinson's disease (PD). Thanks to its transduction properties, Engrailed has been shown to confer neuroprotection in several experimental models of PD. Study of the mechanisms underlying these two Engrailed-mediated effects has revealed a key role of the translation regulation by Engrailed and uncovered an unsuspected link between a homeoprotein and mitochondrial activity. These studies highlight the crucial role of cellular energetic metabolism in neuron development, survival and neurodegeneration, and may help to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Julia Fuchs
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), CNRS unité mixte de recherche 7241/INSERM U1050, Development and Neuropharmacology, 11 place Marcelin Berthelot, Paris F-75005, France
| | | | | | | | | | | |
Collapse
|
23
|
Lo C, Flinn LJ, Bandmann O. Heterozygous mutations in the FGF8, SHH and nodal/transforming growth factor beta pathways do not confer increased dopaminergic neuron vulnerability--a zebrafish study. Neurosci Lett 2012; 532:55-8. [PMID: 23123778 DOI: 10.1016/j.neulet.2012.10.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 10/15/2012] [Accepted: 10/17/2012] [Indexed: 11/29/2022]
Abstract
Fibroblast growth factor 8 (FGF8), sonic hedgehog (SHH) and nodal signalling pathways play key roles in both development and survival of dopaminergic neurons. Both heterozygous mutations in autosomal recessively inherited Parkinson's disease (PD) genes such as parkin or PINK1 and exposure to exogenous toxins are thought to contribute to the pathogenesis of PD. The aim of our study was to investigate whether heterozygote mutations in fgf8, shh or oep lead to a reduced number of ascending dopaminergic neurons in zebrafish (Danio rerio) or confer increased susceptibility to the PD neurotoxin 1-methyl-4-phenyl-pyridinium (MPP⁺). At 3 days post fertilization, heterozygous mutations in fgf8, shh or oep did not affect the number of ascending dopaminergic neurons, nor did heterozygous mutations in fgf8, shh or oep result in increased susceptibility to MPP⁺. Further work is needed to determine whether haploinsufficiency in other neurodevelopmental genes might confer increased susceptibility to PD-related pathomechanisms.
Collapse
Affiliation(s)
- Christine Lo
- Centre for Developmental and Biomedical Genetics-CDBG, Sheffield, UK
| | | | | |
Collapse
|
24
|
Xia YP, Dai RL, Li YN, Mao L, Xue YM, He QW, Huang M, Huang Y, Mei YW, Hu B. The protective effect of sonic hedgehog is mediated by the propidium iodide 3-kinase/AKT/Bcl-2 pathway in cultured rat astrocytes under oxidative stress. Neuroscience 2012; 209:1-11. [DOI: 10.1016/j.neuroscience.2012.02.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 02/12/2012] [Accepted: 02/13/2012] [Indexed: 11/24/2022]
|
25
|
Al-Ayadhi LY. Relationship between Sonic hedgehog protein, brain-derived neurotrophic factor and oxidative stress in autism spectrum disorders. Neurochem Res 2012; 37:394-400. [PMID: 21984201 PMCID: PMC3264868 DOI: 10.1007/s11064-011-0624-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 09/27/2011] [Accepted: 09/29/2011] [Indexed: 10/26/2022]
Abstract
The etiology of autism spectrum disorders (ASD) is not well known but oxidative stress has been suggested to play a pathological role. We report here that the serum levels of Sonic hedgehog (SHH) protein and brain-derived neurotrophic factor (BDNF) might be linked to oxidative stress in ASD. By using the whole blood or polymorphonuclear leukocytes, we demonstrated that autistic children produced a significantly higher level of oxygen free radicals (OFR). In addition, we found significantly higher levels of serum SHH protein in children with mild as well as severe form of autism. We also found that the serum level of BDNF was significantly reduced in autistic children with mild form of the disorder but not with severe form of the disorder. Our findings are the first to report a correlation between SHH, BDNF and OFR in autistic children, suggesting a pathological role of oxidative stress and SHH in autism spectrum disorders.
Collapse
Affiliation(s)
- Laila Y Al-Ayadhi
- Department of Physiology, Autism Research & Treatment Center (ART Center), AL-Amodi Autism Research Chair, Faculty of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia.
| |
Collapse
|
26
|
Peterson R, Turnbull J. Sonic hedgehog is cytoprotective against oxidative challenge in a cellular model of amyotrophic lateral sclerosis. J Mol Neurosci 2011; 47:31-41. [PMID: 21979788 DOI: 10.1007/s12031-011-9660-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 09/26/2011] [Indexed: 01/08/2023]
Abstract
We have previously demonstrated that primary cilia on spinal motor neurons are reduced in G93A SOD1 (mSOD) mice, a mouse model of amyotrophic lateral sclerosis (ALS). Sonic hedgehog (Shh) signaling involves the primary cilium and Shh has been shown to be cytoprotective in models of other neurodegenerative diseases. Thus, the Shh signaling pathway may bear further study in ALS. Accordingly, we established that interference with the Shh pathway (with the Shh antagonist cyclopamine or with miRNA 3245p) sensitized HT22 cells, while augmentation of the Shh pathway (with Shh or the Shh agonist purmorphamine) protected cells against hydrogen peroxide (H₂O₂) challenge. We ectopically expressed mSOD, human wild-type SOD1 (wtSOD), or an empty vector in HT22 cells. Compared to empty vector, wtSOD decreased cell death and mSOD increased cell death in response to H₂O₂ challenge. Treatment with cyclopamine or miRNA 3245p sensitized all three transfections to H₂O₂ challenge. Treatment with recombinant human Shh or purmorphamine decreased cell death after H₂O₂ challenge, an effect more pronounced in mSOD cells. Compared with empty vector, overexpression of wtSOD increased Shh and Gli transcript levels and increased activity in a Gli-responsive reporter assay. Overexpression of mSOD did not change Shh transcript levels, but decreased Gli transcript levels, especially Gli3, and reduced activity in a Gli reporter assay. These results suggest that overexpression of mSOD but not wtSOD reduces signaling in the Shh pathway and renders mSOD cells more susceptible to H₂O₂ challenge, and that treatment with Shh or Shh agonists is cytoprotective to mSOD cells. Shh or Shh agonists merit further consideration as potential therapy in ALS.
Collapse
Affiliation(s)
- Randy Peterson
- Department of Medicine, McMaster University Medical Centre, Rm 4U7, 1200 Main St West, Hamilton, Ontario L8N 3Z5, Canada
| | | |
Collapse
|
27
|
Sonic Hedgehog Protects Cortical Neurons Against Oxidative Stress. Neurochem Res 2010; 36:67-75. [DOI: 10.1007/s11064-010-0264-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2010] [Indexed: 12/28/2022]
|
28
|
Abstract
The discovery of a Sonic Hedgehog (Shh) signaling pathway in the mature vertebrate CNS has paved the way to the characterization of the functional roles of Shh signals in normal and diseased brain. Shh is proposed to participate in the establishment and maintenance of adult neurogenic niches and to regulate the proliferation of neuronal or glial precursors in several brain areas. Consistent with its role during brain development, misregulation of Shh signaling is associated with tumorigenesis while its recruitement in damaged neural tissue might be part of the regenerating process. This review focuses on the most recent data of the Hedgehog pathway in the adult brain and its relevance as a novel therapeutic approach for brain diseases including brain tumors.
Collapse
Affiliation(s)
- Elisabeth Traiffort
- CNRS, Alfred Fessard Institute of Neurobiology, Laboratory of Neurobiology and Development, UPR-3294, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France.
| | | | | |
Collapse
|
29
|
Shinozaki S, Ohnishi H, Hama K, Kita H, Yamamoto H, Osawa H, Sato K, Tamada K, Mashima H, Sugano K. Indian hedgehog promotes the migration of rat activated pancreatic stellate cells by increasing membrane type-1 matrix metalloproteinase on the plasma membrane. J Cell Physiol 2008; 216:38-46. [PMID: 18286538 DOI: 10.1002/jcp.21372] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Indian hedgehog (Ihh) is a member of hedgehog peptides family that exerts diverse effects on multiple cellular functions. Since Ihh expression is elevated in the pancreas of chronic pancreatitis patients, Ihh has been assumed to participate in the chronic pancreatic injury, especially in pancreatic fibrosis. However, its function in pancreatic fibrosis is still unknown. We thus examined Ihh effects on rat activated pancreatic stellate cells (PSCs) that play a central role in pancreatic fibrosis. Activated PSCs express both patched-1 and smoothened that are essential components of hedgehog receptor system. Ihh did not alter the PSC expression of collagen-1 or alpha-smooth muscle actin, a parameter of PSC transformation, or did not change PSC proliferation. However, Ihh enhanced PSC migration in both chemotactic and chemokinetic manners. Furthermore, Ihh increased the amount of membrane-type 1 matrix metalloproteinase (MT1-MMP) and altered its localization on the plasma membrane, which plays a stimulatory role in cellular migration. In addition, tissue inhibitor of metalloproteinase-2 (TIMP-2) attenuated Ihh-stimulated PSC migration. Since most hedgehog intracellular signals are mediated by Gli-1 transcription factor, we investigated its contribution to Ihh-enhancement of PSC migration. Ihh induced Gli-1 nuclear accumulation in PSCs, indicating that Ihh stimulates Gli-1-dependent signaling pathway in PSCs. Unexpectedly, however, adenovirus-mediated Gli-1 overexpression blocked the Ihh enhancement of both MT1-MMP localization on the plasma membrane and PSC migration. Furthermore, reduction of Gli-1 expression with RNA interference augmented Ihh-stimulated PSC migration. These data indicate that Ihh promotes PSC migration by enhancing MT1-MMP localization on the plasma membrane but is negatively regulated by Gli-1.
Collapse
Affiliation(s)
- Satoshi Shinozaki
- Division of Gastroenterology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Carlson ME, Silva HS, Conboy IM. Aging of signal transduction pathways, and pathology. Exp Cell Res 2008; 314:1951-61. [PMID: 18474281 DOI: 10.1016/j.yexcr.2008.03.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 03/26/2008] [Accepted: 03/29/2008] [Indexed: 12/15/2022]
Abstract
The major cell signaling pathways, and their specific mechanisms of transduction, have been a subject of investigation for many years. As our understanding of these pathways advances, we find that they are evolutionarily well-conserved not only individually, but also at the level of their crosstalk and signal integration. Productive interactions within the key signal transduction networks determine success in embryonic organogenesis, and postnatal tissue repair throughout adulthood. However, aside from clues revealed through examining age-related degenerative diseases, much remains uncertain about imbalances within these pathways during normal aging. Further, little is known about the molecular mechanisms by which alterations in the major cell signal transduction networks cause age-related pathologies. The aim of this review is to describe the complex interplay between the Notch, TGFbeta, WNT, RTK-Ras and Hh signaling pathways, with a specific focus on the changes introduced within these networks by the aging process, and those typical of age-associated human pathologies.
Collapse
Affiliation(s)
- Morgan E Carlson
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720-1762, USA.
| | | | | |
Collapse
|
31
|
King GD, Kroeger KM, Bresee CJ, Candolfi M, Liu C, Manalo CM, Muhammad AKMG, Pechnick RN, Lowenstein PR, Castro MG. Flt3L in combination with HSV1-TK-mediated gene therapy reverses brain tumor-induced behavioral deficits. Mol Ther 2008; 16:682-90. [PMID: 18283279 PMCID: PMC2593113 DOI: 10.1038/mt.2008.18] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 01/16/2008] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an invasive and aggressive primary brain tumor which is associated with a dismal prognosis. We have earlier developed a macroscopic, intracranial, syngeneic GBM model, in which treatment with adenoviral vectors (Ads) expressing herpes simplex virus type 1 thymidine kinase (HSV1-TK) plus ganciclovir (GCV) resulted in survival of approximately 20% of the animals. In this model, treatment with Ads expressing Fms-like tyrosine kinase 3 ligand (Flt3L), in combination with Ad-HSV1-TK improves the survival rate to approximately 70% and induces systemic antitumor immunity. We hypothesized that the growth of a large intracranial tumor mass would cause behavioral abnormalities that can be reversed by the combined gene therapy. We assessed the behavior and neuropathology of tumor-bearing animals treated with the combined gene therapy, 3 days after treatment, in long-term survivors, and in a recurrent model of glioma. We demonstrate that the intracranial GBM induces behavioral deficits that are resolved after treatment with Ad-Flt3L/Ad-TK (+GCV). Neuropathological analysis of long-term survivors revealed an overall recovery of normal brain architecture. The lack of long-term behavioral deficits and limited neuropathological abnormalities demonstrate the efficacy and safety of the combined Ad-Flt3L/Ad-TK gene therapy for GBM. These findings can serve to underpin further developments of this therapeutic modality, leading toward implementation of a Phase I clinical trial.
Collapse
Affiliation(s)
- Gwendalyn D King
- Board of Governors' Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Cytotoxic exposure of bone marrow and other non-hematopoietic organs containing self-renewing stem cell populations is associated with damage to the supportive microenvironment. Recent evidence indicates that radical oxygen species resulting from the initial oxidative stress persist for months after ionizing irradiation exposure of tissues including oral cavity, esophagus, lung and bone marrow. Antioxidant gene therapy using manganese superoxide dismutase plasmid liposomes has provided organ-specific radiation protection associated with delay or prevention of acute and late toxicity. Recent evidence has suggested that manganese superoxide dismutase transgene expression in cells of the organ microenvironment contributes significantly to the mechanism of protection. Incorporating this knowledge into designs of novel approaches for stem cell protection is addressed in the present review.
Collapse
Affiliation(s)
- J S Greenberger
- Department of Radiation Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213-2532, USA.
| |
Collapse
|
33
|
Zeng Z, Yuan X, Liu G, Zeng X, Zeng X, Ng H, Chen H, Jiang T, Akasaki Y, Kessey K, Black KL, Yu JS. Manipulation of proliferation and differentiation of human bone marrow-derived neural stem cells in vitro and in vivo. J Neurosci Res 2007; 85:310-20. [PMID: 17131390 DOI: 10.1002/jnr.21131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Recent evidence has demonstrated that neural stem cells (NSC) can be expanded from a variety of sources, including embryos, fetuses, and adult bone marrow and brain tissue. We have previously reported the generation of adult rat bone marrow-derived cellular spheres that are morphologically and phenotypically similar to neurospheres derived from brain NSC. Here we show that adult human bone marrow-derived neural stem cells (HBM-NSC) are capable of generating spheres that are similar to brain neural-derived neurospheres. Additionally, we sought to promote proliferation and differentiation of HBM-NSC through transduction with nonreplicative recombinant adenovirus encoding the cDNA sequence for Gli, rADV-Gli-1; sonic hedgehog, rADV-Shh; or Nurr1, rADV-Nurr1. Immunocytochemistry and RT-PCR analysis showed that HBM-NSC could be efficiently expanded and differentiated in vitro and that HBM-NSC transduced with rADV-Gli-1 or rADV-Shh dramatically increased NSC time-related proliferation; however, Nurr1 had no effect on proliferation. We also transplanted HBM-NSC into chicken embryos to examine their potential function in vivo. We found that transduction of HBM-NSC with rADV-Gli-1 or rADV-Shh and subsequent transplantation into chicken embryos increased HBM-NSC proliferation, whereas rADV-Nurr1 promoted migration and differentiation in vivo. Our findings suggest that HBM-NSC can be efficiently expanded and differentiated in vitro and in vivo by overexpressing Gli-1, Shh or Nurr1.
Collapse
Affiliation(s)
- Zhaohui Zeng
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kan I, Melamed E, Offen D. Autotransplantation of bone marrow-derived stem cells as a therapy for neurodegenerative diseases. Handb Exp Pharmacol 2007:219-42. [PMID: 17554511 DOI: 10.1007/978-3-540-68976-8_10] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurodegenerative diseases are characterized by a progressive degeneration of selective neural populations. This selective hallmark pathology and the lack of effective treatment modalities make these diseases appropriate candidates for cell therapy. Bone marrow-derived mesenchymal stem cells (MSCs) are self-renewing precursors that reside in the bone marrow and may further be exploited for autologous transplantation. Autologous transplantation of MSCs entirely circumvents the problem of immune rejection, does not cause the formation of teratomas, and raises very few ethical or political concerns. More than a few studies showed that transplantation of MSCs resulted in clinical improvement. However, the exact mechanisms responsible for the beneficial outcome have yet to be defined. Possible rationalizations include cell replacement, trophic factors delivery, and immunomodulation. Cell replacement theory is based on the idea that replacement of degenerated neural cells with alternative functioning cells induces long-lasting clinical improvement. It is reasoned that the transplanted cells survive, integrate into the endogenous neural network, and lead to functional improvement. Trophic factor delivery presents a more practical short-term approach. According to this approach, MSC effectiveness may be credited to the production of neurotrophic factors that support neuronal cell survival, induce endogenous cell proliferation, and promote nerve fiber regeneration at sites of injury. The third potential mechanism of action is supported by the recent reports claiming that neuroinflammatory mechanisms play an important role in the pathogenesis of neurodegenerative disorders. Thus, inhibiting chronic inflammatory stress might explain the beneficial effects induced by MSC transplantation. Here, we assemble evidence that supports each theory and review the latest studies that have placed MSC transplantation into the spotlight of biomedical research.
Collapse
Affiliation(s)
- I Kan
- Laboratory of Neurosciences, Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Campus Tel Aviv University, Sackler School of Medicine, 49100 Petah-Tikva, Israel
| | | | | |
Collapse
|
35
|
Barcia C, Gerdes C, Xiong WD, Thomas CE, Liu C, Kroeger KM, Castro MG, Lowenstein PR. Immunological thresholds in neurological gene therapy: highly efficient elimination of transduced cells might be related to the specific formation of immunological synapses between T cells and virus-infected brain cells. NEURON GLIA BIOLOGY 2006; 2:309-22. [PMID: 18084640 PMCID: PMC2139984 DOI: 10.1017/s1740925x07000579] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
First-generation adenovirus can be engineered with powerful promoters to drive expression of therapeutic transgenes. Numerous clinical trials for glioblastoma multiforme using first generation adenoviral vectors have either been performed or are ongoing, including an ongoing, Phase III, multicenter trial in Europe and Israel (Ark Therapeutics, Inc.). Although in the absence of anti-adenovirus immune responses expression in the brain lasts 6-18 months, systemic infection with adenovirus induces immune responses that inhibit dramatically therapeutic transgene expression from first generation adenoviral vectors, thus, potentially compromising therapeutic efficacy. Here, we show evidence of an immunization threshold for the dose that generates an immune response strong enough to eliminate transgene expression from the CNS. For the systemic immunization to eliminate transgene expression from the brain, > or = 1 x 10(7) infectious units (iu) of adenovirus need to be used as immunogen. Furthermore, this immune response eliminates >90% of transgene expression from 1 x 10(7)-1 x 10(3) iu of vector injected into the striatum 60 days earlier. Importantly, elimination of transgene expression is independent of the nature of the promoter that drives transgene expression and is accompanied by brain infiltration of CD8(+) T cells and macrophages. In conclusion, once the threshold for systemic immunization (i.e. 1 x 10(7) iu) is crossed, the immune response eliminates transgene expression by >90% even from brains that receive as little as 1000 iu of adenoviral vectors, independently of the type of promoter that drives expression.
Collapse
Affiliation(s)
- Carlos Barcia
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 5094, Los Angeles, USA
| | - Christian Gerdes
- Molecular Medicine and Gene Therapy Unit, Room 1.302, Stopford Building, University of Manchester, UK
| | - Wei-Dong Xiong
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 5094, Los Angeles, USA
| | - Clare E. Thomas
- Molecular Medicine and Gene Therapy Unit, Room 1.302, Stopford Building, University of Manchester, UK
| | - Chunyan Liu
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 5094, Los Angeles, USA
| | - Kurt M. Kroeger
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 5094, Los Angeles, USA
| | - Maria G. Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 5094, Los Angeles, USA
- Departments of Medicine and Molecular and Medical Pharmacology, David Gerfen School of Medicine, University of California Los Angeles, USA
| | - Pedro R. Lowenstein
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Room 5094, Los Angeles, USA
- Departments of Medicine and Molecular and Medical Pharmacology, David Gerfen School of Medicine, University of California Los Angeles, USA
| |
Collapse
|
36
|
PUNTEL M, CURTIN J, ZIRGER J, MUHAMMAD A, XIONG W, LIU C, HU J, KROEGER K, CZER P, SCIASCIA S, MONDKAR S, LOWENSTEIN P, CASTRO M. Quantification of high-capacity helper-dependent adenoviral vector genomes in vitro and in vivo, using quantitative TaqMan real-time polymerase chain reaction. Hum Gene Ther 2006; 17:531-44. [PMID: 16716110 PMCID: PMC1592228 DOI: 10.1089/hum.2006.17.531] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
First-generation adenoviral (Ad) and high-capacity adenoviral (HC-Ad) vectors are efficient delivery vehicles for transferring therapeutic transgenes in vivo into tissues/organs. The initial successes reported with adenoviral vectors in preclinical trials have been limited by immune-related adverse side effects. This has been, in part, attributed to the use of poorly characterized preparations of adenoviral vectors and also to the untoward immune adverse side effects elicited when high doses of these vectors were used. HC-Ads have several advantages over Ads, including the lack of viral coding sequences, which after infection and uncoating, makes them invisible to the host's immune system. Another advantage is their large cloning capacity (up to approximately 35 kb). However, accurate characterization of HC-Ad vectors, and of contaminating replication-competent adenovirus (RCA) or helper virus, is necessary before these preparations can be used safely in clinical trials. Consequently, the development of accurate, simple, and reproducible methods to standardize and validate adenoviral preparations for the presence of contaminant genomes is required. By using a molecular method that allows accurate, reproducible, and simultaneous determination of HC-Ad, contaminating helper virus, and RCA genome copy numbers based on real-time quantitative PCR, we demonstrate accurate detection of these three genomic entities, within CsCl-purified vector stocks, total DNA isolated from cells transduced in vitro, and from brain tissue infected in vivo. This approach will allow accurate assessment of the levels and biodistribution of HC-Ad and improve the safety and efficacy of clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - P.R. LOWENSTEIN
- Address reprint requests to: Dr. M.G. Castro or Dr. P.R. Lowenstein, Board of Governors’ Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Davis Building, Research Pavilion, Room 5090, Los Angeles, CA 90048, E-mail:or
| | - M.G. CASTRO
- Address reprint requests to: Dr. M.G. Castro or Dr. P.R. Lowenstein, Board of Governors’ Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Davis Building, Research Pavilion, Room 5090, Los Angeles, CA 90048, E-mail:or
| |
Collapse
|
37
|
Puntel M, Curtin J, Zirger J, Muhammad A, Xiong W, Liu C, Hu J, Kroeger K, Czer P, Sciascia S, Mondkar S, Lowenstein P, Castro M. Quantification of High-Capacity Helper-Dependent Adenoviral Vector Genomes In Vitro and In Vivo, Using Quantitative TaqMan Real-Time Polymerase Chain Reaction. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
38
|
Osawa H, Ohnishi H, Takano K, Noguti T, Mashima H, Hoshino H, Kita H, Sato K, Matsui H, Sugano K. Sonic hedgehog stimulates the proliferation of rat gastric mucosal cells through ERK activation by elevating intracellular calcium concentration. Biochem Biophys Res Commun 2006; 344:680-7. [PMID: 16630542 DOI: 10.1016/j.bbrc.2006.03.188] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2006] [Accepted: 03/28/2006] [Indexed: 12/20/2022]
Abstract
Sonic Hedgehog (Shh), a member of hedgehog peptides family, is expressed in gastric gland epithelium. To elucidate Shh function to gastric mucosal cells, we examined the effect of Shh on the proliferation of a rat normal gastric mucosal cell line, RGM-1. RGM-1 cells express essential components of Shh receptor system, patched-1, and smoothened. Shh enhanced DNA synthesis in RGM-1 cells and elevated intracellular calcium concentration ([Ca2+]i). In addition, Shh as well as calcium ionophore A32187 rapidly activated ERK. However, Shh failed to activate ERK under calcium-free culture condition. Pretreatment of cells with PD98059 attenuated the DNA synthesis promoted by Shh. Moreover, when cells were pretreated with cyclopamine, Shh could not elevate [Ca2+]i, activate ERK or promote DNA synthesis. On the other hand, although Shh induced Gli-1 nuclear accumulation in RGM-1 cells, Shh activated ERK even in cells pretreated with actinomycin D. These results indicate that Shh promotes the proliferation of RGM-1 cells through an intracellular calcium- and ERK-dependent but transcription-independent pathway via Patched/Smoothened receptor system.
Collapse
Affiliation(s)
- Hiroyuki Osawa
- Department of Gastroenterology, Jichi Medical School, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Jacobs AH, Winkler A, Castro MG, Lowenstein P. Human gene therapy and imaging in neurological diseases. Eur J Nucl Med Mol Imaging 2006; 32 Suppl 2:S358-83. [PMID: 16328505 PMCID: PMC2902257 DOI: 10.1007/s00259-005-1960-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Molecular imaging aims to assess non-invasively disease-specific biological and molecular processes in animal models and humans in vivo. Apart from precise anatomical localisation and quantification, the most intriguing advantage of such imaging is the opportunity it provides to investigate the time course (dynamics) of disease-specific molecular events in the intact organism. Further, molecular imaging can be used to address basic scientific questions, e.g. transcriptional regulation, signal transduction or protein/protein interaction, and will be essential in developing treatment strategies based on gene therapy. Most importantly, molecular imaging is a key technology in translational research, helping to develop experimental protocols which may later be applied to human patients. Over the past 20 years, imaging based on positron emission tomography (PET) and magnetic resonance imaging (MRI) has been employed for the assessment and "phenotyping" of various neurological diseases, including cerebral ischaemia, neurodegeneration and brain gliomas. While in the past neuro-anatomical studies had to be performed post mortem, molecular imaging has ushered in the era of in vivo functional neuro-anatomy by allowing neuroscience to image structure, function, metabolism and molecular processes of the central nervous system in vivo in both health and disease. Recently, PET and MRI have been successfully utilised together in the non-invasive assessment of gene transfer and gene therapy in humans. To assess the efficiency of gene transfer, the same markers are being used in animals and humans, and have been applied for phenotyping human disease. Here, we review the imaging hallmarks of focal and disseminated neurological diseases, such as cerebral ischaemia, neurodegeneration and glioblastoma multiforme, as well as the attempts to translate gene therapy's experimental knowledge into clinical applications and the way in which this process is being promoted through the use of novel imaging approaches.
Collapse
Affiliation(s)
- Andreas H Jacobs
- Max Planck-Institute for Neurological Research, Center of Molecular Medicine (CMMC) and Department of Neurology, University of Cologne, Cologne, Germany.
| | | | | | | |
Collapse
|
40
|
Torres EM, Monville C, Lowenstein PR, Castro MG, Dunnett SB. Delivery of sonic hedgehog or glial derived neurotrophic factor to dopamine-rich grafts in a rat model of Parkinson's disease using adenoviral vectors Increased yield of dopamine cells is dependent on embryonic donor age. Brain Res Bull 2005; 68:31-41. [PMID: 16325002 PMCID: PMC2902250 DOI: 10.1016/j.brainresbull.2005.08.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The poor survival of dopamine grafts in Parkinson's disease is one of the main obstacles to the widespread application of this therapy. One hypothesis is that implanted neurons, once removed from the embryonic environment, lack the differentiation factors needed to develop the dopaminergic phenotype. In an effort to improve the numbers of dopamine neurons surviving in the grafts, we have investigated the potential of adenoviral vectors to deliver the differentiation factor sonic hedgehog or the glial cell line-derived neurotrophic factor GDNF to dopamine-rich grafts in a rat model of Parkinson's disease. Adenoviral vectors containing sonic hedgehog, GDNF, or the marker gene LacZ were injected into the dopamine depleted striatum of hemiparkinsonian rats. Two weeks later, ventral mesencephalic cell suspensions were prepared from embryos of donor ages E12, E13, E14 or E15 and implanted into the vector-transduced striatum. Pre-treatment with the sonic hedgehog vector produced a three-fold increase in the numbers of tyrosine hydroxylase-positive (presumed dopaminergic) cells in grafts derived from E12 donors, but had no effect on E13-E15 grafts. By contrast, pre-treatment with the GDNF vector increased yields of dopamine cells in grafts derived from E14 and E15 donors but had no effect on grafts from younger donors. The results indicate that provision of both trophic and differentiation factors can enhance the yields of dopamine neurons in ventral mesencephalic grafts, but that the two factors differ in the age and stage of embryonic development at which they have maximal effects.
Collapse
Affiliation(s)
- E M Torres
- Department of Biosciences, Cardiff University, Biomedical Sciences Building, Museum Avenue, PO Box 911, Cardiff CF10 3US, UK.
| | | | | | | | | |
Collapse
|
41
|
Suwelack D, Hurtado-Lorenzo A, Millan E, Gonzalez-Nicolini V, Wawrowsky K, Lowenstein PR, Castro MG. Neuronal expression of the transcription factor Gli1 using the Talpha1 alpha-tubulin promoter is neuroprotective in an experimental model of Parkinson's disease. Gene Ther 2005; 11:1742-52. [PMID: 15573088 PMCID: PMC1249480 DOI: 10.1038/sj.gt.3302377] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nigrostriatal neurons degenerate during Parkinson's disease. Experimentally, neurotoxins such as 6-hydroxydopamine (6-OHDA) in rodents, and MPTP in mice and non-human primates, are used to model the disease-induced degeneration of midbrain dopaminergic neurons. Glial-cell-derived neurotrophic factor (GDNF) is a very powerful neuroprotector of dopaminergic neurons in all species examined. However, recent reports have indicated the possibility that GDNF may, in the long term and if expressed in an unregulated manner, exert untoward effects on midbrain dopaminergic neuronal structure and function. Although GDNF remains a powerful neurotrophin, the search for alternative therapies based on alternative and complementary mechanisms of action to GDNF is warranted. Recently, recombinant adenovirus-derived vectors encoding the differentiation factor Sonic Hedgehog (Shh) and its downstream transcriptional activator (Gli1) were shown to protect dopaminergic neurons in the substantia nigra pars compacta from 6-OHDA-induced neurotoxicity in rats in vivo. A pancellular human CMV (hCMV) promoter was used to drive the expression of both Shh and Gli1. Since Gli1 is a transcription factor and therefore exerts its actions intracellularly, we decided to test whether expression of Gli1 within neurons would be effective for neuroprotection. We demonstrate that neuronal-specific expression of Gli1 using the neuron-specific Talpha1 alpha-tubulin (Talpha1) promoter was neuroprotective, and its efficiency was comparable to the pancellular strong viral hCMV promoter. These results suggest that expression of the transcription factor Gli1 solely within neurons is neuroprotective for dopaminergic neurons in vivo and, furthermore, that neuronal-specific promoters are effective within the context of adenovirus-mediated gene therapy-induced neuroprotection of dopaminergic midbrain neurons. Since cell-type specific promoters are known to be weaker than the viral hCMV promoter, our data demonstrate that neuronal-specific expression of transcription factors is an effective, specific, and sufficient targeted approach for neurological gene therapy applications, potentially minimizing side effects due to unrestricted promiscuous gene expression within target tissues.
Collapse
Affiliation(s)
- D Suwelack
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | |
Collapse
|