1
|
Xu J, Liu D, Xiao S, Meng X, Zhao D, Jiang X, Jiang X, Cai L, Jiang H. Low-Dose Radiation Prevents Chemotherapy-Induced Cardiotoxicity. CURRENT STEM CELL REPORTS 2019; 5:82-91. [DOI: 10.1007/s40778-019-00158-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
2
|
Abstract
AbstractIn recent decades, clinical trials in Alzheimer’s disease (AD) have failed at an unprecedented rate. The etiology of AD has since come under renewed scrutiny, both to elucidate the underlying pathologies and to identify novel therapeutic strategies. Here, diet has emerged as a potential causative/protective agent. A variety of nutrients, including lipids, minerals, vitamins, antioxidants and sugars as well as broader dietary patterns and microbiotal interactions have demonstrated associations with AD. Although clinical trials have yet to definitively implicate any singular dietary element as therapeutic or causative, it is apparent that dietary preferences, likely in complex synergies, may influence the risk, onset and course of AD. This review catalogs the impact of major dietary elements on AD. It further examines an unexplored reciprocal association where AD may modulate diet, as well as how potential therapeutics may complicate these interactions. In doing so, we observe diet may have profound effects on the outcome of a clinical trial, either as a confounder of a drug/disease interaction or as a generally disruptive covariate. We therefore conclude that future clinical trials in AD should endeavor to control for diet, either in study design or subsequent analyses.
Collapse
|
3
|
The interplay between histone deacetylases and rho kinases is important for cancer and neurodegeneration. Cytokine Growth Factor Rev 2017; 37:29-45. [PMID: 28606734 DOI: 10.1016/j.cytogfr.2017.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/18/2017] [Accepted: 05/21/2017] [Indexed: 12/24/2022]
Abstract
Rho associated coiled-coil containing kinases (ROCKs) respond to defined extra- and intracellular stimuli to control cell migration, cell proliferation, and apoptosis. Histone deacetylases (HDACs) are epigenetic modifiers that regulate nuclear and cytoplasmic signaling through the deacetylation of histones and non-histone proteins. ROCK and HDAC functions are important compounds of basic and applied research interests. Recent evidence suggests a physiologically important interplay between HDACs and ROCKs in various cells and organisms. Here we summarize the crosstalk between these enzymatic families and its implications for cancer and neurodegeneration.
Collapse
|
4
|
Seo Y, Kim HS, Kang I, Choi SW, Shin TH, Shin JH, Lee BC, Lee JY, Kim JJ, Kook MG, Kang KS. Cathepsin S contributes to microglia-mediated olfactory dysfunction through the regulation of Cx3cl1-Cx3cr1 axis in a Niemann-Pick disease type C1 model. Glia 2016; 64:2291-2305. [PMID: 27687148 DOI: 10.1002/glia.23077] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 12/28/2022]
Abstract
Microglia can aggravate olfactory dysfunction by mediating neuronal death in the olfactory bulb (OB) of a murine model of Niemann-Pick disease type C1 (NPC1), a fatal neurodegenerative disorder accompanied by lipid trafficking defects. In this study, we focused on the crosstalk between neurons and microglia to elucidate the mechanisms underlying extensive microgliosis in the NPC1-affected brain. Microglia in the OB of NPC1 mice strongly expressed CX3C chemokine receptor 1 (Cx3cr1), a specific receptor for the neural chemokine C-X3-C motif ligand 1 (Cx3cl1). In addition, a high level of Cx3cl1 was detected in NPC1 mouse-derived CSF due to enhanced catalytic activity of Cathepsin S (Ctss), which is responsible for Cx3cl1 secretion. Notably, nasal delivery of Cx3cl1 neutralizing antibody or Ctss inhibitor could inhibit the Cx3cl1-Cx3cr1 interaction and support neuronal survival through the suppression of microglial activation, leading to an improvement in the olfactory function in NPC1 mice. Relevant in vitro experiments revealed that intracellular cholesterol accumulation could act as a strong inducer of abnormal Ctss activation and, in turn, stimulated the Cx3cl1-Cx3cr1 axis in microglia via p38 mitogen-activated protein kinase signaling. Our data address the significance of Cx3cl1-Cx3cr1 interaction in the development of microglial neurotoxicity and suggest that Ctss is a key upstream regulator. Therefore, this study contributes to a better understanding of the crosstalk between neurons and microglia in the development of the neurodegeneration and provides a new perspective for the management of olfactory deficits and other microglia-dependent neuropathies. GLIA 2016;64:2291-2305.
Collapse
Affiliation(s)
- Yoojin Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,School of Medicine, Pusan National University, Busan, 49241, South Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, South Korea
| | - Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,School of Medicine, Pusan National University, Busan, 49241, South Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, South Korea
| | - Insung Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Tae-Hoon Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Ji-Hee Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Byung-Chul Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jin Young Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jae-Jun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Myung Geun Kook
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea. .,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
5
|
Network Topology Analysis of Post-Mortem Brain Microarrays Identifies More Alzheimer's Related Genes and MicroRNAs and Points to Novel Routes for Fighting with the Disease. PLoS One 2016; 11:e0144052. [PMID: 26784894 PMCID: PMC4718516 DOI: 10.1371/journal.pone.0144052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 11/12/2015] [Indexed: 12/17/2022] Open
Abstract
Network-based approaches are powerful and beneficial tools to study complex systems in their entirety, elucidating the essential factors that turn the multitude of individual elements into a functional system. In this study we used critical network topology descriptors and guilt-by-association rule to explore and understand the significant molecular players, drug targets and underlying biological mechanisms of Alzheimer’s disease. Analyzing two post-mortem brain gene microarrays (GSE4757 and GSE28146) with Pathway Studio software package we constructed and analyzed a set of protein-protein interaction, as well as miRNA-target networks. In a 4-step procedure the expression datasets were normalized using Robust Multi-array Average approach, while the modulation of gene expression by the disease was statistically evaluated by the empirical Bayes method from the limma Bioconductor package. Representative set of 214 seed-genes (p<0.01) common for the three brain sections of the two microarrays was thus created. The Pathway Studio analysis of the networks built identified 15 new potential AD-related genes and 17 novel AD-involved microRNAs. Using KEGG pathways relevant in Alzheimer’s disease we built an integrated mechanistic network from the interactions between the overlapping genes in these pathways. Routes of possible disease initiation process were thus revealed through the CD4, DCN, and IL8 extracellular ligands. DAVID and IPA enrichment analysis uncovered a number of deregulated biological processes and pathways including neuron projection/differentiation, aging, oxidative stress, chemokine/ neurotrophin signaling, long-term potentiation and others. The findings in this study offer information of interest for subsequent experimental studies.
Collapse
|
6
|
Zhang JG, Zhang DD, Wu X, Wang YZ, Gu SY, Zhu GH, Li XY, Li Q, Liu GL. Incarvine C suppresses proliferation and vasculogenic mimicry of hepatocellular carcinoma cells via targeting ROCK inhibition. BMC Cancer 2015; 15:814. [PMID: 26510899 PMCID: PMC4625643 DOI: 10.1186/s12885-015-1809-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023] Open
Abstract
Background Studies have described vasculogenic mimicry (VM) as an alternative circulatory system to blood vessels in multiple malignant tumor types, including hepatocellular carcinoma (HCC). In the current study, we aimed to seek novel and more efficient treatment strategies by targeting VM and explore the underlying mechanisms in HCC cells. Methods Cell counting kit-8 (CCK-8) assay and colony survival assay were performed to explore the inhibitory effect of incarvine C (IVC) on human cancer cell proliferation. Flow cytometry was performed to analyze the cell cycle distribution after DNA staining and cell apoptosis by the Annexin V-PE and 7-AAD assay. The effect of IVC on Rho-associated, coiled-coil-containing protein kinase (ROCK) was determined by western blotting and stress fiber formation assay. The inhibitory role of IVC on MHCC97H cell VM formation was determined by formation of tubular network structures on Matrigel in vitro, real time-qPCR, confocal microscopy and western blotting techniques. Results We explored an anti-metastatic HCC agent, IVC, derived from traditional Chinese medicinal herbs, and found that IVC dose-dependently inhibited the growth of MHCC97H cells. IVC induced MHCC97H cell cycle arrest at G1 transition, which was associated with cyclin-dependent kinase 2 (CDK-2)/cyclin-E1 degradation and p21/p53 up-regulation. In addition, IVC induced apoptotic death of MHCC97H cells. Furthermore, IVC strongly suppressed the phosphorylation of the ROCK substrate myosin phosphatase target subunit-1 (MYPT-1) and ROCK-mediated actin fiber formation. Finally, IVC inhibited cell-dominant tube formation in vitro, which was accompanied with the down-regulation of VM-key factors as detected by real time-qPCR and immunofluorescence. Conclusions Taken together, the effective inhibitory effect of IVC on MHCC97H cell proliferation and neovascularization was associated with ROCK inhibition, suggesting that IVC may be a new potential drug candidate for the treatment of HCC.
Collapse
Affiliation(s)
- Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Dan-Dan Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Xin Wu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Yu-Zhu Wang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Sheng-Ying Gu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Guan-Hua Zhu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Xiao-Yu Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Qin Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| | - Gao-Lin Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of medicine, No. 100 Haining Road, Shanghai, 200080, P. R. China.
| |
Collapse
|
7
|
Abstract
Dendritic protein synthesis and actin cytoskeleton reorganization are important events required for the consolidation of hippocampal LTP and memory. However, the temporal and spatial relationships between these two processes remain unclear. Here, we report that treatment of adult rat hippocampal slices with BDNF or with tetraethylammonium (TEA), which induces a chemical form of LTP, produces a rapid and transient increase in RhoA protein levels. Changes in RhoA were restricted to dendritic spines of CA3 and CA1 and require de novo protein synthesis regulated by mammalian target of rapamycin (mTOR). BDNF-mediated stimulation of RhoA activity, cofilin phosphorylation, and actin polymerization were completely suppressed by protein synthesis inhibitors. Furthermore, intrahippocampal injections of RhoA antisense oligodeoxynucleotides inhibited theta burst stimulation (TBS)-induced RhoA upregulation in dendritic spines and prevented LTP consolidation. Addition of calpain inhibitors after BDNF or TEA treatment maintained RhoA levels elevated and prolonged the effects of BDNF and TEA on actin polymerization. Finally, the use of isoform-selective calpain inhibitors revealed that calpain-2 was involved in RhoA synthesis, whereas calpain-1 mediated RhoA degradation. Overall, this mechanism provides a novel link between dendritic protein synthesis and reorganization of the actin cytoskeleton in hippocampal dendritic spines during LTP consolidation.
Collapse
|
8
|
Liao G, Wang Z, Lee E, Moreno S, Abuelnasr O, Baudry M, Bi X. Enhanced expression of matrix metalloproteinase-12 contributes to Npc1 deficiency-induced axonal degeneration. Exp Neurol 2015; 269:67-74. [PMID: 25864931 DOI: 10.1016/j.expneurol.2015.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 01/08/2015] [Accepted: 04/02/2015] [Indexed: 11/28/2022]
Abstract
Niemann-Pick type C (NPC) disease is a genetic disorder associated with intracellular cholesterol accumulation in the brain and other organs, and neurodegeneration is generally believed to be the fatal cause of the disease. In view of the emerging role of matrix metalloproteinase-12 (MMP-12) in neuronal injury, we investigated its expression and potential roles in axonal degeneration in Npc1-/- mouse brain. Microarray and quantitative real-time reversed transcription PCR analysis indicated a marked increase in MMP-12 mRNA levels in cerebellum of 3 week-old Npc1-/- mice, as compared to wild-type littermates. Western blots showed that the ratio of mature MMP-12 over pro-MMP-12 was significantly increased in cerebellum of Npc1-/-, as compared to wild-type mice. Immunohistochemical studies confirmed that MMP-12 expression was increased, especially in the cell bodies of Purkinje neurons in Npc1-/- mice. Neuritic growth was significantly reduced by Npc1 siRNA knockdown in nerve growth factor-differentiated PC-12 cells, and this effect was completely reversed by treatment with an MMP-12 specific inhibitor. Furthermore, in vivo experiments showed that chronic treatment with the MMP-12 inhibitor ameliorated Npc1 deficiency-induced axonal pathology in the striatum. Our results indicate that abnormal neuronal expression of MMP-12 may contribute to axonal degeneration in NPC disease, thus providing a potential novel target for treatment.
Collapse
Affiliation(s)
- Guanghong Liao
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Zhuangjun Wang
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Erik Lee
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Stephanie Moreno
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Omar Abuelnasr
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA
| | - Xiaoning Bi
- Department of Basic Medical Sciences, COMP, Western University of Health Sciences, 701 E. Second Street, Pomona, CA 91766, USA.
| |
Collapse
|
9
|
Mar FM, da Silva TF, Morgado MM, Rodrigues LG, Rodrigues D, Pereira MIL, Marques A, Sousa VF, Coentro J, Sá-Miranda C, Sousa MM, Brites P. Myelin Lipids Inhibit Axon Regeneration Following Spinal Cord Injury: a Novel Perspective for Therapy. Mol Neurobiol 2015; 53:1052-1064. [PMID: 25579385 DOI: 10.1007/s12035-014-9072-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/29/2014] [Indexed: 11/28/2022]
Abstract
Lack of axon regeneration following spinal cord injury has been mainly ascribed to the inhibitory environment of the injury site, i.e., to chondroitin sulfate proteoglycans (CSPGs) and myelin-associated inhibitors (MAIs). Here, we used shiverer (shi) mice to assess axon regeneration following spinal cord injury in the presence of MAIs and CSPG but in the absence of compact myelin. Although in vitro shi neurons displayed a similar intrinsic neurite outgrowth to wild-type neurons, in vivo, shi fibers had increased regenerative capacity, suggesting that the wild-type spinal cord contains additional inhibitors besides MAIs and CSPG. Our data show that besides myelin protein, myelin lipids are highly inhibitory for neurite outgrowth and suggest that this inhibitory effect is released in the shi spinal cord given its decreased lipid content. Specifically, we identified cholesterol and sphingomyelin as novel myelin-associated inhibitors that operate through a Rho-dependent mechanism and have inhibitory activity in multiple neuron types. We further demonstrated the inhibitory action of myelin lipids in vivo, by showing that delivery of 2-hydroxypropyl-β-cyclodextrin, a drug that reduces the levels of lipids specifically in the injury site, leads to increased axon regeneration of wild-type (WT) dorsal column axons following spinal cord injury. In summary, our work shows that myelin lipids are important modulators of axon regeneration that should be considered together with protein MAIs as critical targets in strategies aiming at improving axonal growth following injury.
Collapse
Affiliation(s)
- Fernando M Mar
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Tiago F da Silva
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Marlene M Morgado
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | - Lorena G Rodrigues
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.,Lysosome and Peroxisome Biology group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | - Daniel Rodrigues
- Lysosome and Peroxisome Biology group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | - Marta I L Pereira
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | - Ana Marques
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | - Vera F Sousa
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - João Coentro
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | - Clara Sá-Miranda
- Lysosome and Peroxisome Biology group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| | - Mónica M Sousa
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.
| | - Pedro Brites
- Nerve Regeneration group, Instituto de Biologia Molecular e Celular-IBMC, University of Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal
| |
Collapse
|
10
|
Yan X, Yang F, Lukas J, Witt M, Wree A, Rolfs A, Luo J. Hyperactive glial cells contribute to axonal pathologies in the spinal cord of Npc1 mutant mice. Glia 2014; 62:1024-40. [PMID: 24644136 DOI: 10.1002/glia.22659] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/12/2014] [Accepted: 03/04/2014] [Indexed: 12/27/2022]
Abstract
Niemann-Pick disease type C1 (NPC1) is a neurodegenerative disease with various progressive pathological features, for example, neuronal loss, dysmyelination, abnormal axon swelling, and gliosis, in the brain. Pathological activation of p38-mitogen-activated protein kinase (MAPK) results in hyperphosphorylation of tau protein, which contributes to the development of neurodegenerative diseases. In this study, axonal varicosities or spheroids and presynaptic aggregates in the spinal cord of the Npc1 mutant mice were found from postnatal day (P) 35 onwards, as indicated by the increased hyperphosphorylated neurofilament and synaptophysin immunoreactivity as well as the findings from electron microscopy. However, activities of astrocytes and microglia in the Npc1 mutant spinal cord were progressively increased earlier from P10 onwards, accompanied by increased expression of interleukin-1β and apolipoprotein E, as well as up-regulated p38-MAPK activity and enhanced phosphorylated tau protein, but not cyclin-dependent kinase 5/p35 complex and glycogen synthase kinase-3β. Taken together, our data suggest that the axonal pathologies in the Npc1 mutant spinal cord are strongly correlated with the increase of activated glial cells, which produce IL-1β and ApoE, resulting in the activation of p38-MAPK signaling pathway and enhanced phosphorylated tau protein.
Collapse
Affiliation(s)
- Xin Yan
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | |
Collapse
|
11
|
Sun J, Sun G, Meng X, Wang H, Luo Y, Qin M, Ma B, Wang M, Cai D, Guo P, Sun X. Isorhamnetin protects against doxorubicin-induced cardiotoxicity in vivo and in vitro. PLoS One 2013; 8:e64526. [PMID: 23724057 PMCID: PMC3665796 DOI: 10.1371/journal.pone.0064526] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 04/15/2013] [Indexed: 12/21/2022] Open
Abstract
Doxorubicin (Dox) is an anthracycline antibiotic for cancer therapy with limited usage due to cardiotoxicity. Isorhamnetin is a nature antioxidant with obvious cardiac protective effect. The aim of this study is going to investigate the possible protective effect of isorhamnetin against Dox-induced cardiotoxicity and its underlying mechanisms. In an in vivo investigation, rats were intraperitoneally (i.p.) administered with Dox to duplicate the model of Dox-induced chronic cardiotoxicity. Daily pretreatment with isorhamnetin (5 mg/kg, i.p.) for 7 days was found to reduce Dox-induced myocardial damage significantly, including the decline of cardiac index, decrease in the release of serum cardiac enzymes and amelioration of heart vacuolation. In vitro studies on H9c2 cardiomyocytes, isorhamnetin was effective to reduce Dox-induced cell toxicity. A further mechanism study indicated that isorhamnetin pretreatment can counteract Dox-induced oxidative stress and suppress the activation of mitochondrion apoptotic pathway and mitogen-activated protein kinase pathway. Isorhamnetin also potentiated the anti-cancer activity of Dox in MCF-7, HepG2 and Hep2 cells. These findings indicated that isorhamnetin can be used as an adjuvant therapy for the long-term clinical use of Dox.
Collapse
Affiliation(s)
- Jing Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Guibo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xiangbao Meng
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Hongwei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Yun Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Meng Qin
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Bo Ma
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Min Wang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Dayong Cai
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Peng Guo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
12
|
Baudry M, Bi X. Learning and memory: an emergent property of cell motility. Neurobiol Learn Mem 2013; 104:64-72. [PMID: 23707799 DOI: 10.1016/j.nlm.2013.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 04/29/2013] [Accepted: 04/30/2013] [Indexed: 01/24/2023]
Abstract
In this review, we develop the argument that the molecular/cellular mechanisms underlying learning and memory are an adaptation of the mechanisms used by all cells to regulate cell motility. Neuronal plasticity and more specifically synaptic plasticity are widely recognized as the processes by which information is stored in neuronal networks engaged during the acquisition of information. Evidence accumulated over the last 25 years regarding the molecular events underlying synaptic plasticity at excitatory synapses has shown the remarkable convergence between those events and those taking place in cells undergoing migration in response to extracellular signals. We further develop the thesis that the calcium-dependent protease, calpain, which we postulated over 25 years ago to play a critical role in learning and memory, plays a central role in the regulation of both cell motility and synaptic plasticity. The findings discussed in this review illustrate the general principle that fundamental cell biological processes are used for a wide range of functions at the level of organisms.
Collapse
Affiliation(s)
- Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA.
| | | |
Collapse
|
13
|
Abstract
Cholesterol is an essential membrane component enriched in plasma membranes, growth cones, and synapses. The brain normally synthesizes all cholesterol locally, but the contribution of individual cell types to brain cholesterol metabolism is unknown. To investigate whether cortical projection neurons in vivo essentially require cholesterol biosynthesis and which cell types support neurons, we have conditionally ablated the cholesterol biosynthesis in these neurons in mice either embryonically or postnatally. We found that cortical projection neurons synthesize cholesterol during their entire lifetime. At all stages, they can also benefit from glial support. Adult neurons that lack cholesterol biosynthesis are mainly supported by astrocytes such that their functional integrity is preserved. In contrast, microglial cells support young neurons. However, compensatory efforts of microglia are only transient leading to layer-specific neuronal death and the reduction of cortical projections. Hence, during the phase of maximal membrane growth and maximal cholesterol demand, neuronal cholesterol biosynthesis is indispensable. Analysis of primary neurons revealed that neurons tolerate only slight alteration in the cholesterol content and plasma membrane tension. This quality control allows neurons to differentiate normally and adjusts the extent of neurite outgrowth, the number of functional growth cones and synapses to the available cholesterol. This study highlights both the flexibility and the limits of horizontal cholesterol transfer in vivo and may have implications for the understanding of neurodegenerative diseases.
Collapse
|
14
|
Ma J, Xue Y, Cui W, Li Y, Zhao Q, Ye W, Zheng J, Cheng Y, Ma Y, Li S, Han T, Miao L, Yao L, Zhang J, Liu W. Ras homolog gene family, member A promotes p53 degradation and vascular endothelial growth factor-dependent angiogenesis through an interaction with murine double minute 2 under hypoxic conditions. Cancer 2012; 118:4105-16. [PMID: 22907703 DOI: 10.1002/cncr.27393] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 11/15/2011] [Accepted: 11/28/2011] [Indexed: 01/15/2023]
Abstract
BACKGROUND Tumor neovascularization (TNV) is a common pathologic basis for malignant growth and metastasis. However, the mechanism of TNV pathogenesis is not fully understood. Ras homolog gene family, member A (RhoA), a Rho guanosine triphosphatase (GTPase) family member, may be involved in a hypoxia-induced vascular endothelial growth factor (VEGF) pathway that regulates TNV angiogenesis through an unclear mechanism. METHODS The regulation of RhoA on p53, the p53 binding protein homolog murine double minute 2 (MDM2), and VEGF was analyzed in hypoxic MCF-7 cells using Western blot analysis, real-time polymerase chain reaction (PCR) analysis, coimmunoprecipitation, and immunofluorescence staining assays. Changes in proliferation, invasion, migration, stress fiber formation, and tube formation were detected in an MCF-7 human umbilical vein endothelial cell (HUVEC) coculture system. Correlations of RhoA expression with MDM2, wild-type p53 (wt-p53), and VEGF expression in breast cancer tissues and relations between RhoA and breast cancer clinical features were analyzed by immunohistochemistry. RESULTS Activated RhoA down-regulated p53 protein, which increased VEGF expression in hypoxic MCF-7 cells; whereas p53 messenger RNA levels were not altered. In addition, the ubiquitin-mediated degradation of p53 was enhanced by active RhoA. RhoA and MDM2 colocalized in the cytoplasm of hypoxic MCF-7 cells and interacted with each other physically. Furthermore, nutlin-3, a specific MDM2 inhibitor, was capable of reducing activated RhoA-induced p53 protein stability and attenuating VEGF accumulation. In an MCF-7-HUVEC coculture system, nutlin-3 effectively inhibited HUVEC proliferation, invasion, migration, stress fiber formation, and tube formation mediated by activated RhoA under hypoxic conditions. Data from 129 clinical breast cancer specimens with wt-p53 revealed that high RhoA expression was correlated with high MDM2 expression, low wt-p53 expression, and high VEGF expression. CONCLUSIONS The current data suggested that activated RhoA promotes VEGF expression and hypoxia-induced angiogenesis through the up-regulation of MDM2 to decrease p53 stability.
Collapse
Affiliation(s)
- Ji Ma
- Department of Oncology, Xijing Hospital, State Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yang G, Qu X, Zhang J, Zhao W, Wang H. Sema3F downregulates p53 expression leading to axonal growth cone collapse in primary hippocampal neurons. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2012; 5:634-41. [PMID: 22977659 PMCID: PMC3438774 DOI: pmid/22977659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 07/26/2012] [Indexed: 11/18/2022]
Abstract
Hippocampal nerve growth is regulated by the coordinated action of numerous external stimuli, including positively acting neurotrophin-derived growth cues and restrictive semaphorin cues, however the underlying cellular mechanisms remain largely unclear. We examined the potential cellular mechanism of Semaphorin3F (Sema3F) in cultured primary hippocampal neurons. We show that Sema3F can down-regulate p53 expression in primary hippocampal neurons, thereby contributing to growth cone collapse. Sema3F suppressed p53-induced pathways, which we show to be required to maintain growth cone structure. Sema3F-induced growth cone collapse was partially reversed by overexpression of p53, which promoted growth cone extension. Inhibition of p53 function by inhibitor, siRNAs, induced axonal growth cone collapse, whereas p53 over-expression led to larger growth cones in cultured primary hippocampal neurons.These data reveal a novel mechanism by which Sema3F can induce hippocampal neuron growth cone collapse and provide evidence for an intracellular mechanism for cross talk between positive and negative axon growth cues.
Collapse
Affiliation(s)
- Guanglu Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | | | | | | | | |
Collapse
|
16
|
Street CA, Bryan BA. Rho kinase proteins--pleiotropic modulators of cell survival and apoptosis. Anticancer Res 2011; 31:3645-3657. [PMID: 22110183 PMCID: PMC3226732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Rho kinase (ROCK) proteins are Rho-GTPase activated serine/threonine kinases that function as modulators of actin-myosin cytoskeletal dynamics via regulation of Lin11, Isl-1 & Mec-3 domain (LIM) kinase, myosin light chain (MLC), and MLC phosphatase. A strong correlation between cytoskeletal rearrangements and tumor cell invasion, metastasis, and deregulated microenvironment interaction has been reported in the literature, and the utilization of pharmacological inhibitors of ROCK signaling for the treatment of cancer is actively being pursued by a number of pharmaceutical companies. Indeed, in many preclinical models ROCK inhibitors have shown remarkable efficacy in reducing tumor growth and metastasis. Interestingly, ROCK signaling has been shown to be either pro-apoptotic or pro-survival in a cell type and context dependent manner, though the molecular mechanisms controlling ROCK-mediated cell fate decisions are unknown. This review summarizes the many pleiotropic roles of ROCK signaling in survival and apoptosis, and suggests that controlled modulation of ROCK activity in tumor cells has the potential to significantly affect tumor survival and patient outcome.
Collapse
Affiliation(s)
- Catharine A. Street
- Ghosh Science and Technology Center, Department of Biology, Worcester State University, Worcester, MA, 01602, U.S.A
| | - Brad A. Bryan
- Center of Excellence in Cancer Research, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, U.S.A
| |
Collapse
|
17
|
Zadran S, Bi X, Baudry M. Regulation of calpain-2 in neurons: implications for synaptic plasticity. Mol Neurobiol 2010; 42:143-50. [PMID: 20924799 DOI: 10.1007/s12035-010-8145-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2010] [Accepted: 09/26/2010] [Indexed: 12/28/2022]
Abstract
The family of calcium-dependent neutral proteases, calpains, was discovered more than 30 years ago, but their functional roles in the nervous system under physiological or pathological conditions still remain unclear. Although calpain was proposed to participate in synaptic plasticity and in learning and memory in the early 1980s, the precise mechanism regarding its activation, its target(s) and the functional consequences of its activation have remained controversial. A major issue has been the identification of roles of the two major calpain isoforms present in the brain, calpain-1 and calpain-2, and the calcium requirement for their activation, which exceeds levels that could be reached intracellularly under conditions leading to changes in synaptic efficacy. In this review, we discussed the features of calpains that make them ideally suited to link certain patterns of presynaptic activity to the structural modifications of dendritic spines that could underlie synaptic plasticity and learning and memory. We then summarize recent findings that provide critical answers to the various questions raised by the initial hypothesis, and that further support the idea that, in brain, calpain-2 plays critical roles in developmental and adult synaptic plasticity.
Collapse
Affiliation(s)
- Sohila Zadran
- Neuroscience Program, University of Southern California, Los Angeles, CA 90089-2520, USA
| | | | | |
Collapse
|
18
|
Role of calpain-mediated p53 truncation in semaphorin 3A-induced axonal growth regulation. Proc Natl Acad Sci U S A 2010; 107:13883-7. [PMID: 20643936 DOI: 10.1073/pnas.1008652107] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Neurite outgrowth represents a critical stage in the correct development of neuronal circuitries, and is dependent on the complex regulation of actin filament and microtubule dynamics by intrinsic as well as extrinsic signals. Previous studies have implicated the tumor suppressor factor, p53, in the regulation of axonal outgrowth through a nontranscriptional effect involving local regulation of the Rho kinase signaling pathway that controls these dynamics. In the present study, we first showed that semaphorin 3A-induced growth cone collapse in cultured hippocampal neurons was associated with the partial truncation of phosphorylated p53, and that both effects were prevented by calpain inhibition with either m-calpain-specific siRNA or inhibitors. We further determined that semaphorin 3A-mediated calpain activation and growth cone collapse were associated with m-calpain phosphorylation and prevented by inhibition of MAPK, ERK, or p38. In vitro studies confirmed that p53 and especially phosphorylated p53 were partially truncated by calpain. Thus, our results indicate that semaphorin 3A-mediated growth cone collapse is mediated in part by m-calpain activation, possibly through MAPK-mediated phosphorylation, and the resulting truncation of phosphorylated p53, leading to Rho kinase activation and cytoskeletal reorganization. They provide a pathway by which extrinsic signals regulate axonal growth through activation of m-calpain and p53 truncation.
Collapse
|