1
|
Pehar M, Hewitt M, Wagner A, Sandhu JK, Khalili A, Wang X, Cho JY, Sim VL, Kulka M. Histamine stimulates human microglia to alter cellular prion protein expression via the HRH2 histamine receptor. Sci Rep 2024; 14:25519. [PMID: 39462031 PMCID: PMC11513956 DOI: 10.1038/s41598-024-75982-1] [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: 06/05/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Although the cellular prion protein (PrPC) has been evolutionarily conserved, the role of this protein remains elusive. Recent evidence indicates that PrPC may be involved in neuroinflammation and the immune response in the brain, and its expression may be modified via various mechanisms. Histamine is a proinflammatory mediator and neurotransmitter that stimulates numerous cells via interactions with histamine receptors 1-4 (HRH1-4). Since microglia are the innate immune cells of the central nervous system, we hypothesized that histamine-induced stimulation regulates the expression of PrPC in human-derived microglia. The human microglial clone 3 (HMC3) cell line was treated with histamine, and intracellular calcium levels were measured via a calcium flux assay. Cytokine production was monitored by enzyme-linked immunosorbent assay (ELISA). Western blotting and quantitative reverse transcription-polymerase chain reaction were used to determine protein and gene expression of HRH1-4. Flow cytometry and western blotting were used to measure PrPC expression levels. Fluorescence microscopy was used to examine Iba-1 and PrPC localization. HMC3 cells stimulated by histamine exhibited increased intracellular calcium levels and increased release of IL-6 and IL-8, while also modifying PrPC localization. HMC3 stimulated with histamine for 6 and 24 hours exhibited increased surface PrPC expression. Specifically, we found that stimulation of the HRH2 receptor was responsible for changes in surface PrPC. Histamine-induced increases in surface PrPC were attenuated following inhibition of the HRH2 receptor via the HRH2 antagonist ranitidine. These changes were unique to HRH2 activation, as stimulation of HRH1, HRH3, or HRH4 did not alter surface PrPC. Prolonged stimulation of HMC3 decreased PrPC expression following 48 and 72 hours of histamine stimulation. HMC3 cells can be stimulated by histamine to undergo intracellular calcium influx. Surface expression levels of PrPC on HMC3 cells are altered by histamine exposure, primarily mediated by HRH2. While histamine exposure also increases release of IL-6 and IL-8 in these cells, this cytokine release is not fully dependent on PrPC levels, as IL-6 release is only partially reduced and IL-8 release is unchanged under the conditions of HRH2 blockade that prevent PrPC changes. Overall, this suggests that PrPC may play a role in modulating microglial responses.
Collapse
Affiliation(s)
- Marcus Pehar
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Melissa Hewitt
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Ashley Wagner
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
| | - Jagdeep K Sandhu
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Aria Khalili
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Xinyu Wang
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Jae-Young Cho
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Valerie L Sim
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Marianna Kulka
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, Edmonton, AB, Canada.
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
2
|
Xu M, Shi R, Yang J, Chen H, Liu S, Yu S, Li S, He W, Sy MS, Lu M, Zhang H, Li C. Collagen prolyl 4-hydroxylase subunit α member-induced head and neck squamous cell carcinoma aggressiveness is antagonized by LLGL2 via reduced expression of occludin. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39394821 DOI: 10.3724/abbs.2024140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024] Open
Abstract
There are three isoforms of human collagen prolyl 4-hydroxylases (C-P4Hs), each of which has been reported to play an important role in regulating the progression of a variety of human cancers. By analyzing TGCA datasets on human head and neck squamous cell carcinoma (HNSC), we find that a higher expression of all three C-P4HAs (the α subunit of C-P4Hs) is a superior prognostic indicator than a higher expression of two or a single C-P4HA. Unexpectedly, some patients with higher levels of three C-P4HAs survive longer than patients whose tumors have lower expression of C-P4HAs. Therefore, there may be molecule(s) that can negate the deleterious effects of overexpressing C-P4HAs during cancer progression. By constructing a functional protein interaction network of C-P4HAs and analyzing molecules whose expressions are correlated significantly with that of C-P4HAs, we identify scribble cell polarity complex component 2 (LLGL2) as a factor that antagonizes the effects of overexpressed C-P4HAs on HNSC. Silencing of LLGL2 in the human oral squamous cell line Cal-27 upregulates the expression of occludin and increases cancer cell invasion and migration. In contrast, knocking down C-P4HA alone inhibits cell migration and invasion. Furthermore, simultaneously downregulating three C-P4HAs has more pronounced effects on inhibiting cell migration and invasion. Accordingly, high LLGL2 expression is also a marker indicating improved prognosis in patients with HNSC. These results suggest that the interplay between LLGL2 and C-P4HAs may be targeted to mitigate HNSC tumorigenesis and progression.
Collapse
Affiliation(s)
- Miao Xu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, School of Basic Medical Sciences, University of South China, Hengyang 421001, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Run Shi
- School of Medicine, Pingdingshan University, Pingdingshan 467000, China
| | - Jie Yang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Heng Chen
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou 510095, China
| | - Shihua Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shupei Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Sasa Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenqiang He
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Mingjian Lu
- Department of Interventional Radiology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Huixia Zhang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chaoyang Li
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, School of Basic Medical Sciences, University of South China, Hengyang 421001, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou 510095, China
| |
Collapse
|
3
|
Liu Y, Zhang J, Zhai Z, Liu C, Yang S, Zhou Y, Zeng X, Liu J, Zhang X, Nie X, Xu J, Huang J, Liu C, Liu Z, Guo M, Sun G. Upregulated PrP C by HBx enhances NF-κB signal via liquid-liquid phase separation to advance liver cancer. NPJ Precis Oncol 2024; 8:211. [PMID: 39333690 PMCID: PMC11437096 DOI: 10.1038/s41698-024-00697-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024] Open
Abstract
Cellular prion protein (PrPC) has been implicated in carcinogenic through the activation of various signal pathways, however, the precise mechanisms remain elusive. In vitro studies have shown that PrPC is prone to undergo liquid-liquid phase separation (LLPS). However, it remains unknown whether PrPC contributes to LLPS-inducing cancer development. Herein, we observed an upregulation of PrPC expression in hepatitis B virus-positive hepatocellular carcinoma (HCC). Subsequent investigation revealed that HBx of HBV enhances PrPC expression in a dose-dependent manner by binding to CREB1. Furthermore, we demonstrated that PrPC undergoes LLPS and recruits TRAF2/6, TAB2/3, and TAK1 protein, thereby activating the NF-κB signaling pathway and promoting tumor progression. Notably, although unable to undergo LLPS itself, the α3 helix of PrPC is essential for its activation of the NF-κB signaling pathway during the LLPS process. Further analysis unveiled that disulfide bond formation within the C-terminal domain of PrPC is necessary for its LLPS and subsequent activation of the NF-κB signaling pathway. Additionally, our findings indicate that NF-κB activation by PrPC condensates leads to increased IL-8 expression which further promotes tumor development.
Collapse
Affiliation(s)
- Yang Liu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jing Zhang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Zixu Zhai
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Chenyi Liu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Siqi Yang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Ying Zhou
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Xianhuang Zeng
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jiaqi Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Xiaoyu Zhang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Xinqi Nie
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jiaqi Xu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Junsong Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Chaozhi Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Zhepeng Liu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China
| | - Mingxiong Guo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China.
- School of Ecology and Environment, Tibet University, Lhasa, 850000, Tibet, People's Republic of China.
| | - Guihong Sun
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, People's Republic of China.
- Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
4
|
Karner D, Kvestak D, Kucan Brlic P, Cokaric Brdovcak M, Lisnic B, Brizic I, Juranic Lisnic V, Golemac M, Tomac J, Krmpotic A, Karkeni E, Libri V, Mella S, Legname G, Altmeppen HC, Hasan M, Jonjic S, Lenac Rovis T. Prion protein alters viral control and enhances pathology after perinatal cytomegalovirus infection. Nat Commun 2024; 15:7754. [PMID: 39237588 PMCID: PMC11377837 DOI: 10.1038/s41467-024-51931-4] [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: 09/20/2023] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
Cytomegalovirus (CMV) infection poses risks to newborns, necessitating effective therapies. Given that the damage includes both viral infection of brain cells and immune system-related damage, here we investigate the involvement of cellular prion protein (PrP), which plays vital roles in neuroprotection and immune regulation. Using a murine model, we show the role of PrP in tempering neonatal T cell immunity during CMV infection. PrP-null mice exhibit enhanced viral control through elevated virus-specific CD8 T cell responses, leading to reduced viral titers and pathology. We further unravel the molecular mechanisms by showing CMV-induced upregulation followed by release of PrP via the metalloproteinase ADAM10, impairing CD8 T cell response specifically in neonates. Additionally, we confirm PrP downregulation in human CMV (HCMV)-infected fibroblasts, underscoring the broader relevance of our observations beyond the murine model. Furthermore, our study highlights how PrP, under the stress of viral pathogenesis, reveals its impact on neonatal immune modulation.
Collapse
Affiliation(s)
- Dubravka Karner
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Daria Kvestak
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Paola Kucan Brlic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | | | - Berislav Lisnic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Ilija Brizic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Vanda Juranic Lisnic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Mijo Golemac
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Tomac
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Astrid Krmpotic
- Department of Histology and Embryology; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Esma Karkeni
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Valentina Libri
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Sebastien Mella
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Giuseppe Legname
- Department of Neuroscience, Prion Biology Laboratory, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Milena Hasan
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS); Institut Pasteur, Université Paris Cité, Paris, France
| | - Stipan Jonjic
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia
| | - Tihana Lenac Rovis
- Center for Proteomics; Faculty of Medicine; University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
5
|
Assis-de-Lemos G, Moura-do-Nascimento R, Amaral-do-Nascimento M, Miceli AC, Vieira TCRG. Interactions between Cytokines and the Pathogenesis of Prion Diseases: Insights and Implications. Brain Sci 2024; 14:413. [PMID: 38790392 PMCID: PMC11117815 DOI: 10.3390/brainsci14050413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/26/2024] Open
Abstract
Transmissible Spongiform Encephalopathies (TSEs), including prion diseases such as Bovine Spongiform Encephalopathy (Mad Cow Disease) and variant Creutzfeldt-Jakob Disease, pose unique challenges to the scientific and medical communities due to their infectious nature, neurodegenerative effects, and the absence of a cure. Central to the progression of TSEs is the conversion of the normal cellular prion protein (PrPC) into its infectious scrapie form (PrPSc), leading to neurodegeneration through a complex interplay involving the immune system. This review elucidates the current understanding of the immune response in prion diseases, emphasizing the dual role of the immune system in both propagating and mitigating the disease through mechanisms such as glial activation, cytokine release, and blood-brain barrier dynamics. We highlight the differential cytokine profiles associated with various prion strains and stages of disease, pointing towards the potential for cytokines as biomarkers and therapeutic targets. Immunomodulatory strategies are discussed as promising avenues for mitigating neuroinflammation and delaying disease progression. This comprehensive examination of the immune response in TSEs not only advances our understanding of these enigmatic diseases but also sheds light on broader neuroinflammatory processes, offering hope for future therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Tuane C. R. G. Vieira
- Institute of Medical Biochemistry Leopoldo de Meis and National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (G.A.-d.-L.); (R.M.-d.-N.); (M.A.-d.-N.); (A.C.M.)
| |
Collapse
|
6
|
Risen SJ, Boland SW, Sharma S, Weisman GM, Shirley PM, Latham AS, Hay AJD, Gilberto VS, Hines AD, Brindley S, Brown JM, McGrath S, Chatterjee A, Nagpal P, Moreno JA. Targeting Neuroinflammation by Pharmacologic Downregulation of Inflammatory Pathways Is Neuroprotective in Protein Misfolding Disorders. ACS Chem Neurosci 2024; 15:1533-1547. [PMID: 38507813 DOI: 10.1021/acschemneuro.3c00846] [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] [Indexed: 03/22/2024] Open
Abstract
Neuroinflammation plays a crucial role in the development of neurodegenerative protein misfolding disorders. This category of progressive diseases includes, but is not limited to, Alzheimer's disease, Parkinson's disease, and prion diseases. Shared pathogenesis involves the accumulation of misfolded proteins, chronic neuroinflammation, and synaptic dysfunction, ultimately leading to irreversible neuronal loss, measurable cognitive deficits, and death. Presently, there are few to no effective treatments to halt the advancement of neurodegenerative diseases. We hypothesized that directly targeting neuroinflammation by downregulating the transcription factor, NF-κB, and the inflammasome protein, NLRP3, would be neuroprotective. To achieve this, we used a cocktail of RNA targeting therapeutics (SB_NI_112) shown to be brain-penetrant, nontoxic, and effective inhibitors of both NF-κB and NLRP3. We utilized a mouse-adapted prion strain as a model for neurodegenerative diseases to assess the aggregation of misfolded proteins, glial inflammation, neuronal loss, cognitive deficits, and lifespan. Prion-diseased mice were treated either intraperitoneally or intranasally with SB_NI_112. Behavioral and cognitive deficits were significantly protected by this combination of NF-κB and NLRP3 downregulators. Treatment reduced glial inflammation, protected against neuronal loss, prevented spongiotic change, rescued cognitive deficits, and significantly lengthened the lifespan of prion-diseased mice. We have identified a nontoxic, systemic pharmacologic that downregulates NF-κB and NLRP3, prevents neuronal death, and slows the progression of neurodegenerative diseases. Though mouse models do not always predict human patient success and the study was limited due to sample size and number of dosing methods utilized, these findings serve as a proof of principle for continued translation of the therapeutic SB_NI_112 for prion disease and other neurodegenerative diseases. Based on the success in a murine prion model, we will continue testing SB_NI_112 in a variety of neurodegenerative disease models, including Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Sydney J Risen
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
- Brain Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Sean W Boland
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
- Brain Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Sadhana Sharma
- Sachi Bioworks Inc., Colorado Technology Center, 685 South Arthur Avenue, Louisville, Colorado 80027, United States
| | - Grace M Weisman
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Payton M Shirley
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Amanda S Latham
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Arielle J D Hay
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Vincenzo S Gilberto
- Sachi Bioworks Inc., Colorado Technology Center, 685 South Arthur Avenue, Louisville, Colorado 80027, United States
| | - Amelia D Hines
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Stephen Brindley
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Jared M Brown
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Stephanie McGrath
- Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Anushree Chatterjee
- Sachi Bioworks Inc., Colorado Technology Center, 685 South Arthur Avenue, Louisville, Colorado 80027, United States
| | - Prashant Nagpal
- Sachi Bioworks Inc., Colorado Technology Center, 685 South Arthur Avenue, Louisville, Colorado 80027, United States
| | - Julie A Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
- Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
- Brain Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| |
Collapse
|
7
|
Smeele PH, Vaccari T. Snapshots from within the cell: Novel trafficking and non trafficking functions of Snap29 during tissue morphogenesis. Semin Cell Dev Biol 2023; 133:42-52. [PMID: 35256275 DOI: 10.1016/j.semcdb.2022.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 01/27/2023]
Abstract
Membrane trafficking is a core cellular process that supports diversification of cell shapes and behaviors relevant to morphogenesis during development and in adult organisms. However, how precisely trafficking components regulate specific differentiation programs is incompletely understood. Snap29 is a multifaceted Soluble N-ethylmaleimide-sensitive factor Attachment protein Receptor, involved in a wide range of trafficking and non-trafficking processes in most cells. A body of knowledge, accrued over more than two decades since its discovery, reveals that Snap29 is essential for establishing and maintaining the operation of a number of cellular events that support cell polarity and signaling. In this review, we first summarize established functions of Snap29 and then we focus on novel ones in the context of autophagy, Golgi trafficking and vesicle fusion at the plasma membrane, as well as on non-trafficking activities of Snap29. We further describe emerging evidence regarding the compartmentalisation and regulation of Snap29. Finally, we explore how the loss of distinct functions of human Snap29 may lead to the clinical manifestations of congenital disorders such as CEDNIK syndrome and how altered SNAP29 activity may contribute to the pathogenesis of cancer, viral infection and neurodegenerative diseases.
Collapse
Affiliation(s)
- Paulien H Smeele
- Department of Biosciences, Università Degli Studi Di Milano, Milan, Italy
| | - Thomas Vaccari
- Department of Biosciences, Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
8
|
Huang Z. A Function of Amyloid-β in Mediating Activity-Dependent Axon/Synapse Competition May Unify Its Roles in Brain Physiology and Pathology. J Alzheimers Dis 2023; 92:29-57. [PMID: 36710681 PMCID: PMC10023438 DOI: 10.3233/jad-221042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Amyloid-β protein precursor (AβPP) gives rise to amyloid-β (Aβ), a peptide at the center of Alzheimer's disease (AD). AβPP, however, is also an ancient molecule dating back in evolution to some of the earliest forms of metazoans. This suggests a possible ancestral function that may have been obscured by those that evolve later. Based on literature from the functions of Aβ/AβPP in nervous system development, plasticity, and disease, to those of anti-microbial peptides (AMPs) in bacterial competition as well as mechanisms of cell competition uncovered first by Drosophila genetics, I propose that Aβ/AβPP may be part of an ancient mechanism employed in cell competition, which is subsequently co-opted during evolution for the regulation of activity-dependent neural circuit development and plasticity. This hypothesis is supported by foremost the high similarities of Aβ to AMPs, both of which possess unique, opposite (i.e., trophic versus toxic) activities as monomers and oligomers. A large body of data further suggests that the different Aβ oligomeric isoforms may serve as the protective and punishment signals long predicted to mediate activity-dependent axonal/synaptic competition in the developing nervous system and that the imbalance in their opposite regulation of innate immune and glial cells in the brain may ultimately underpin AD pathogenesis. This hypothesis can not only explain the diverse roles observed of Aβ and AβPP family molecules, but also provide a conceptual framework that can unify current hypotheses on AD. Furthermore, it may explain major clinical observations not accounted for and identify approaches for overcoming shortfalls in AD animal modeling.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
9
|
Li H, Zhang J, Ke JR, Yu Z, Shi R, Gao SS, Li JF, Gao ZX, Ke CS, Han HX, Xu J, Leng Q, Wu GR, Li Y, Tao L, Zhang X, Sy MS, Li C. Pro-prion, as a membrane adaptor protein for E3 ligase c-Cbl, facilitates the ubiquitination of IGF-1R, promoting melanoma metastasis. Cell Rep 2022; 41:111834. [PMID: 36543142 DOI: 10.1016/j.celrep.2022.111834] [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: 03/08/2022] [Revised: 08/13/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
Aberrant activation of receptor tyrosine kinase (RTK) is usually a result of mutation and plays important roles in tumorigenesis. How RTK without mutation affects tumorigenesis remains incompletely understood. Here we show that in human melanomas pro-prion (pro-PrP) is an adaptor protein for an E3 ligase c-Cbl, enabling it to polyubiquitinate activated insulin-like growth factor-1 receptor (IGF-1R), leading to enhanced melanoma metastasis. All human melanoma cell lines studied here express pro-PrP, retaining its glycosylphosphatidylinositol-peptide signal sequence (GPI-PSS). The sequence, PVILLISFLI in the GPI-PSS of pro-PrP, binds c-Cbl, docking c-Cbl to the inner cell membrane, forming a pro-PrP/c-Cbl/IGF-1R trimeric complex. Subsequently, IGF-1R polyubiquitination and degradation are augmented, which increases autophagy and tumor metastasis. Importantly, the synthetic peptide PVILLISFLI disrupts the pro-PrP/c-Cbl/IGF-1R complex, reducing cancer cell autophagy and mitigating tumor aggressiveness in vitro and in vivo. Targeting cancer-associated GPI-PSS may provide a therapeutic approach for treating human cancers expressing pro-PrP.
Collapse
Affiliation(s)
- Huan Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China; Wuhan Institute of Virology, Chinese Academy of Sciences, 44 Xiao Hong Shan Zhong Qu, Wuhan 430030, China
| | - Jie Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Jing-Ru Ke
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ze Yu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Run Shi
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Shan-Shan Gao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Jing-Feng Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Zhen-Xing Gao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Chang-Shu Ke
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Hui-Xia Han
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, No. 1023-1063 Shatai South Road, Guangzhou 510515, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, No. 107 North 2nd Road, Shihezi 832008, China
| | - Qibin Leng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Gui-Ru Wu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China
| | - Yingqiu Li
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, 135 West Xingang Road, Guangzhou 510275, China
| | - Lin Tao
- Department of Pathology, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832008, China
| | - Xianghui Zhang
- Department of Public Health, Shihezi University School of Medicine, Shihezi 832000, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chaoyang Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, 78 Heng Zhi Gang Road, Guangzhou 510095, China.
| |
Collapse
|
10
|
Arshad H, Watts JC. Genetically engineered cellular models of prion propagation. Cell Tissue Res 2022; 392:63-80. [PMID: 35581386 DOI: 10.1007/s00441-022-03630-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/26/2022] [Indexed: 11/02/2022]
Abstract
For over three decades, cultured cells have been a useful tool for dissecting the molecular details of prion replication and the identification of candidate therapeutics for prion disease. A major issue limiting the translatability of these studies has been the inability to reliably propagate disease-relevant, non-mouse strains of prions in cells relevant to prion pathogenesis. In recent years, fueled by advances in gene editing technology, it has become possible to propagate prions from hamsters, cervids, and sheep in immortalized cell lines originating from the central nervous system. In particular, the use of CRISPR-Cas9-mediated gene editing to generate versions of prion-permissive cell lines that lack endogenous PrP expression has provided a blank canvas upon which re-expression of PrP leads to species-matched susceptibility to prion infection. When coupled with the ability to propagate prions in cells or organoids derived from stem cells, these next-generation cellular models should provide an ideal paradigm for identifying small molecules and other biological therapeutics capable of interfering with prion replication in animal and human prion disorders. In this review, we summarize recent advances that have widened the spectrum of prion strains that can be propagated in cultured cells and cutting-edge tissue-based models.
Collapse
Affiliation(s)
- Hamza Arshad
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower Rm. 4KD481, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada.,Department of Biochemistry, University of Toronto, 1 King's College Circle, Medical Sciences Building Rm. 5207, Toronto, ON, M5S 1A8, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower Rm. 4KD481, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada. .,Department of Biochemistry, University of Toronto, 1 King's College Circle, Medical Sciences Building Rm. 5207, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
11
|
Biological Effects of Animal Venoms on the Human Immune System. Toxins (Basel) 2022; 14:toxins14050344. [PMID: 35622591 PMCID: PMC9143185 DOI: 10.3390/toxins14050344] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 02/01/2023] Open
Abstract
Venoms are products of specialized glands and serve many living organisms to immobilize and kill prey, start digestive processes and act as a defense mechanism. Venoms affect different cells, cellular structures and tissues, such as skin, nervous, hematological, digestive, excretory and immune systems, as well as the heart, among other structures. Components of both the innate and adaptive immune systems can be stimulated or suppressed. Studying the effects on the cells and molecules produced by the immune system has been useful in many biomedical fields. The effects of venoms can be the basis for research and development of therapeutic protocols useful in the modulation of the immunological system, including different autoimmune diseases. This review focuses on the understanding of biological effects of diverse venom on the human immune system and how some of their components can be useful for the study and development of immunomodulatory drugs.
Collapse
|
12
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
13
|
Wang Y, Bao G, Zhang M, Xiang J, Zhou H, Wahafu A, Wu W, Ma X, Huo L, Bai X, Xie W, Liu P, Wang M. CRB2 enhances malignancy of glioblastoma via activation of the NF-κB pathway. Exp Cell Res 2022; 414:113077. [DOI: 10.1016/j.yexcr.2022.113077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/04/2022] [Accepted: 02/13/2022] [Indexed: 11/27/2022]
|
14
|
Zou Y, Zhang J, Xu J, Fu L, Xu Y, Wang X, Li Z, Zhu L, Sun H, Zheng H, Guo J. SIRT6 inhibition delays peripheral nerve recovery by suppressing migration, phagocytosis and M2-polarization of macrophages. Cell Biosci 2021; 11:210. [PMID: 34906231 PMCID: PMC8672560 DOI: 10.1186/s13578-021-00725-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Background Silent information regulator 6 (SIRT6) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Prior evidences suggested that the anti-inflammatory function of SIRT6 after spinal cord and brain injury, and it plays a crucial role in macrophages polarization of adipose tissue and skin. However, the role of SIRT6 in macrophages involved peripheral nerve injury is still unknown. Given the prominent role of macrophages in peripheral nerve recovery, we aim to investigate the role of SIRT6 in the regulation of phenotypes shift and functions in macrophages after peripheral nerve injury. Results In the present study, we first identified a significant increase of SIRT6 expression during nerve degeneration and macrophages phagocytosis. Next, we found nerve recovery was delayed after SIRT6 silencing by injected shRNA lentivirus into the crushed sciatic nerve, which exhibited a reduced expression of myelin-related proteins (e.g., MAG and MBP), severer myoatrophy of target muscles, and inferior nerve conduction compared to the shRNA control injected mice. In vitro, we found that SIRT6 inhibition by being treated with a selective inhibitor OSS_128167 or lentivirus transfection impairs migration and phagocytosis capacity of bone marrow-derived macrophages (BMDM). In addition, SIRT6 expression was discovered to be reduced after M1 polarization, but SIRT6 was enhanced after M2 polarization in the monocyte-macrophage cell line RAW264.7 and BMDM. Moreover, SIRT6 inhibition increased M1 macrophage polarization with a concomitant decrease in M2 polarization both in RAW264.7 and BMDM via activating NF-κB and TNF-α expression, and SIRT6 activation by UBCS039 treatment could shift the macrophages from M1 to M2 phenotype. Conclusion Our findings indicate that SIRT6 inhibition impairs peripheral nerve repair through suppressing the migration, phagocytosis, and M2 polarization of macrophages. Therefore, SIRT6 may become a favorable therapeutic target for peripheral nerve injury.
Collapse
Affiliation(s)
- Ying Zou
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Jiaqi Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Jiawei Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Lanya Fu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Yizhou Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China.,Department of Spine Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xianghai Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, China
| | - Zhenlin Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Lixin Zhu
- Department of Spine Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Hao Sun
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
| | - Hui Zheng
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
| | - Jiasong Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China. .,Department of Spine Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China. .,Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Mouillet-Richard S, Ghazi A, Laurent-Puig P. The Cellular Prion Protein and the Hallmarks of Cancer. Cancers (Basel) 2021; 13:cancers13195032. [PMID: 34638517 PMCID: PMC8508458 DOI: 10.3390/cancers13195032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary The cellular prion protein PrPC is best known for its involvement, under its pathogenic isoform, in a group of neurodegenerative diseases. Notwithstanding, an emerging role for PrPC in various cancer-associated processes has attracted increasing attention over recent years. PrPC is overexpressed in diverse types of solid cancers and has been incriminated in various aspects of cancer biology, most notably proliferation, migration, invasion and metastasis, as well as resistance to cytotoxic agents. This article aims to provide a comprehensive overview of the current knowledge of PrPC with respect to the hallmarks of cancer, a reference framework encompassing the major characteristics of cancer cells. Abstract Beyond its causal involvement in a group of neurodegenerative diseases known as Transmissible Spongiform Encephalopathies, the cellular prion protein PrPC is now taking centre stage as an important contributor to cancer progression in various types of solid tumours. The prion cancer research field has progressively expanded in the last few years and has yielded consistent evidence for an involvement of PrPC in cancer cell proliferation, migration and invasion, therapeutic resistance and cancer stem cell properties. Most recent data have uncovered new facets of the biology of PrPC in cancer, ranging from its control on enzymes involved in immune tolerance to its radio-protective activity, by way of promoting angiogenesis. In the present review, we aim to summarise the body of literature dedicated to the study of PrPC in relation to cancer from the perspective of the hallmarks of cancer, the reference framework defined by Hanahan and Weinberg.
Collapse
Affiliation(s)
- Sophie Mouillet-Richard
- Centre de Recherche des Cordeliers, Université de Paris, INSERM, Sorbonne Université, F-75006 Paris, France; (A.G.); (P.L.-P.)
- Correspondence:
| | - Alexandre Ghazi
- Centre de Recherche des Cordeliers, Université de Paris, INSERM, Sorbonne Université, F-75006 Paris, France; (A.G.); (P.L.-P.)
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Université de Paris, INSERM, Sorbonne Université, F-75006 Paris, France; (A.G.); (P.L.-P.)
- Department of Biology, Institut du Cancer Paris CARPEM, APHP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| |
Collapse
|
16
|
Silva JD, Nogueira L, Coelho R, Deus A, Khayat A, Marchi R, Oliveira ED, Santos APD, Cavalli L, Pereira S. HPV-associated penile cancer: Impact of copy number alterations in miRNA/mRNA interactions and potential druggable targets. Cancer Biomark 2021; 32:147-160. [PMID: 34151841 DOI: 10.3233/cbm-210035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Penile cancer (PeCa) is a rare disease, but its incidence has increased worldwide, mostly in HPV+ patients. Nevertheless, there is still no targeted treatment for this carcinoma. OBJECTIVE To predict the main signaling pathways involved in penile tumorigenesis and its potential drug targets. METHODS Genome-wide copy number profiling was performed in 28 PeCa. Integration analysis of CNAs and miRNAs and mRNA targets was performed by DIANA-TarBase v.8. The potential impact of the miRNAs/target genes on biological pathways was assessed by DIANA-miRPath v.3.0. For each miRNA, KEGG pathways were generated based on the tarbase and microT-CDS algorithms. Pharmaco-miR was used to identify associations between miRNAs and their target genes to predict druggable targets. RESULTS 269 miRNAs and 2,395 genes were mapped in cytobands with CNAs. The comparison of the miRNAs mapped at these cytobands and the miRNAs that were predicted to regulate the genes also mapped in these regions, resulted in a set of common 35 miRNAs and 292 genes. Enrichment pathway revealed their involvement in five top signaling pathways. EGFR and COX2 were identified as potential druggable targets. CONCLUSION Our data indicate the potential use of EGFR and COX2 inhibitors as a target treatment for PeCa patients.
Collapse
Affiliation(s)
- Jenilson da Silva
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, MA, Brazil
| | - Leudivan Nogueira
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, MA, Brazil.,Aldenora Bello Cancer Hospital, São Luís, MA, Brazil
| | - Ronald Coelho
- Aldenora Bello Cancer Hospital, São Luís, MA, Brazil
| | - Amanda Deus
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, MA, Brazil.,Aldenora Bello Cancer Hospital, São Luís, MA, Brazil
| | - André Khayat
- Oncology Research Center, Federal University of Pará, Belém, PA, Brazil
| | - Rafael Marchi
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Edivaldo de Oliveira
- Tissue Culture and Cytogenetics Laboratory, Institute of Evandro Chagas, Belém, PA, Brazil
| | - Ana Paula Dos Santos
- Department of Physiological Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Luciane Cavalli
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Silma Pereira
- Laboratory of Genetics and Molecular Biology, Department of Biology, Federal University of Maranhão, São Luís, MA, Brazil
| |
Collapse
|
17
|
Nanoparticle-based fluorescence probe for detection of NF-κB transcription factor in single cell via steric hindrance. Mikrochim Acta 2021; 188:226. [PMID: 34106343 DOI: 10.1007/s00604-021-04878-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
A novel nanoparticle-based fluorescence probe was developed for NF-κB transcription factor detection and in situ imaging via steric hindrance. The probe contains gold nanoparticles (AuNPs) to quench fluorescence, and nucleic acids immobilized on the surface of AuNPs to output fluorescence. In the basal state, Cy5 labeled DNA1 folds its long chain into a hairpin structure and quenches fluorescence by forcing the Cy5 fluorophore close to the surface of AuNPs. After the probe enters the cell, the NF-κB transcription factor can bind to the κB site in the DNA duplex of the nucleic acids. The steric hindrance caused by NF-κB leads to the extension of the long chain of DNA1 and the removal of the Cy5 fluorophore from the surface of AuNPs, thereby restoring the fluorescence of the probe. By measuring NF-κB in cell lysis in vitro, the probe obtains a detection limit of 0.38 nM and the linear range from 0.5 to 16 nM. Repeated measurements showed the recovery in the cell nuclear extract was between 93.38 and 109.32%, with relative standard deviation less than 5%. By monitoring the sub-localization of the Cy5 fluorophore in single cell, the probe system can effectively distinguish active NF-κB (nucleus) and inactive NF-κB (cytoplasm) through in situ imaging. The well-designed probe will make up for the shortcomings of the existing technology, and reveal the regulatory role of transcription factors in many disease processes.
Collapse
|
18
|
Li H, Wang R, Yu Z, Shi R, Zhang J, Gao S, Shao M, Cui S, Gao Z, Xu J, Sy MS, Li C. Tumor Necrosis Factor α Reduces SNAP29 Dependent Autolysosome Formation to Increase Prion Protein Level and Promote Tumor Cell Migration. Virol Sin 2020; 36:458-475. [PMID: 33237393 DOI: 10.1007/s12250-020-00320-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/10/2020] [Indexed: 12/20/2022] Open
Abstract
Tumor Necrosis Factor α (TNFα) is best known as a mediator of inflammation and immunity, and also plays important roles in tumor biology. However, the role of TNFα in tumor biology is complex and not completely understood. In a human melanoma cell line, M2, and a lung carcinoma cell line, A549, TNFα up-regulates prion protein (PrP) level, and promotes tumor cell migration in a PrP dependent manner. Silencing PRNP abrogates TNFα induced tumor cell migration; this phenotype is reversed when PRNP is re-introduced. Treatment with TNFα activates nuclear factor kappa B (NF-κB) signaling, which then mitigates autophagy by reducing the expression of Forkhead Box P3 (FOXP3). Down regulation of FOXP3 reduces the transcription of synaptosome associated protein 29 (SNAP29), which is essential in the fusion of autophagosome and lysosome creating autolysosome. FOXP3 being a bona fide transcription factor for SNAP29 is confirmed in a promoter binding assay. Accordingly, silencing SNAP29 in these cell lines also up-regulates PrP, and promotes tumor cell migration without TNFα treatment. But, when SNAP29 or FOXP3 is silenced in these cells, they are no longer respond to TNFα. Thus, a reduction in autophagy is the underlying mechanism by which expression of PrP is up-regulated, and tumor cell migration is enhanced upon TNFα treatment. Disrupting the TNFα-NF-κB-FOXP3-SNAP29 signaling axis may provide a therapeutic approach to mitigate tumor cell migration.
Collapse
Affiliation(s)
- Huan Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100000, China.,Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Ren Wang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Ze Yu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Run Shi
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Jie Zhang
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, China
| | - Shanshan Gao
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ming Shao
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shuzhong Cui
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China.,Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhenxing Gao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Chaoyang Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China. .,Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 510095, China.
| |
Collapse
|
19
|
Colini Baldeschi A, Vanni S, Zattoni M, Legname G. Novel regulators of PrP C expression as potential therapeutic targets in prion diseases. Expert Opin Ther Targets 2020; 24:759-776. [PMID: 32631090 DOI: 10.1080/14728222.2020.1782384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Prion diseases are rare and fatal neurodegenerative disorders. The key molecular event in these disorders is the misfolding of the physiological form of the cellular prion protein, PrPC, leading to the accumulation of a pathological isoform, PrPSc, with unique features. Both isoforms share the same primary sequence, lacking detectable differences in posttranslational modification, a major hurdle for their biochemical or biophysical independent characterization. The mechanism underlying the conversion of PrPC to PrPSc is not completely understood, so finding an effective therapy to cure prion disorders is extremely challenging. AREAS COVERED This review discusses the strategies for decreasing prion replication and throws a spotlight on the relevance of PrPC in the prion accumulation process. EXPERT OPINION PrPC is the key substrate for prion pathology; hence, the most promising therapeutic approach appears to be the targeting of PrPC to block the production of the infectious isoform. The use of RNA interference and antisense oligonucleotide technologies may offer opportunities for treatment because of their success in clinical trials for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Arianna Colini Baldeschi
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore Di Studi Avanzati (SISSA) , Trieste, Italy
| | - Silvia Vanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per Lo Studio E La Cura Dei Tumori (IRST) IRCCS , Meldola, Italy
| | - Marco Zattoni
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore Di Studi Avanzati (SISSA) , Trieste, Italy
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore Di Studi Avanzati (SISSA) , Trieste, Italy
| |
Collapse
|
20
|
Singh S, Singh TG. Role of Nuclear Factor Kappa B (NF-κB) Signalling in Neurodegenerative Diseases: An Mechanistic Approach. Curr Neuropharmacol 2020; 18:918-935. [PMID: 32031074 PMCID: PMC7709146 DOI: 10.2174/1570159x18666200207120949] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/02/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
A transcriptional regulatory nuclear factor kappa B (NF-κB) protein is a modulator of cellular biological activity via binding to a promoter region in the nucleus and transcribing various protein genes. The recent research implicated the intensive role of nuclear factor kappa B (NF-κB) in diseases like autoimmune disorder, inflammatory, cardiovascular and neurodegenerative diseases. Therefore, targeting the nuclear factor kappa B (NF-κB) protein offers a new opportunity as a therapeutic approach. Activation of IκB kinase/NF-κB signaling pathway leads to the development of various pathological conditions in human beings, such as neurodegenerative, inflammatory disorders, autoimmune diseases, and cancer. Therefore, the transcriptional activity of IκB kinase/NF- κB is strongly regulated at various cascade pathways. The nuclear factor NF-kB pathway plays a major role in the expression of pro-inflammatory genes, including cytokines, chemokines, and adhesion molecules. In response to the diverse stimuli, the cytosolic sequestered NF-κB in an inactivated form by binding with an inhibitor molecule protein (IkB) gets phosphorylated and translocated into the nucleus further transcribing various genes necessary for modifying various cellular functions. The various researches confirmed the role of different family member proteins of NF-κB implicated in expressing various genes products and mediating various cellular cascades. MicroRNAs, as regulators of NF- κB microRNAs play important roles in the regulation of the inflammatory process. Therefore, the inhibitor of NF-κB and its family members plays a novel therapeutic target in preventing various diseases. Regulation of NF- κB signaling pathway may be a safe and effective treatment strategy for various disorders.
Collapse
Affiliation(s)
- Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
21
|
Ke J, Wu G, Zhang J, Li H, Gao S, Shao M, Gao Z, Sy MS, Cao Y, Yang X, Xu J, Li C. Melanoma migration is promoted by prion protein via Akt-hsp27 signaling axis. Biochem Biophys Res Commun 2019; 523:375-381. [PMID: 31870551 DOI: 10.1016/j.bbrc.2019.12.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/07/2019] [Indexed: 10/25/2022]
Abstract
Patients with metastatic melanoma have a poorer prognosis. Prion protein (PrP) in melanoma is known to play an important role in cancer cell migration and invasion by interacting with filamin A (FLNa), a cytolinker protein. To investigate if PrP may contribute to cancer cell mobility independent of its binding to FLNa, we knocked out PRNP in M2 melanoma cell, which lacked FLNa expression. We found that deletion of PRNP in M2 significantly reduced its motility. When PRNP was deleted, the level of Akt was decreased. As a consequence, phosphorylation of small heat shock protein (hsp27) was also reduced, which resulted in polymerization of F-actin rendering the cells less migratory. Accordingly, when PrP was re-expressed in PRNP null M2 cells, the mobility of the recurred cells was rescued, so were the expression levels of Akt and phosphorylated hsp27, resulting in a decrease in the polymerization of F-actin. These results revealed that PrP can play a FLNa independent role in cytoskeletal organization and tumor cell migration by modulating Akt-hsp27-F-actin axis.
Collapse
Affiliation(s)
- Jingru Ke
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, 78 Hengzhigang Road, Guangzhou, 510095, China
| | - Guiru Wu
- The Joint Laboratory for Translational Precision Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan, 430071, China
| | - Jie Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, 78 Hengzhigang Road, Guangzhou, 510095, China; Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, No. 107 North 2nd Road, Shihezi, Xinjiang, 832008, China
| | - Huan Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan, 430071, China
| | - Shanshan Gao
- Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan, 430071, China
| | - Ming Shao
- Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan, 430071, China
| | - Zhenxing Gao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, 78 Hengzhigang Road, Guangzhou, 510095, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Yuchun Cao
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaowen Yang
- Department of the First Abdominal Surgery, Jiangxi Tumor Hospital, Nanchang, Jiangxi, 330029, China.
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, No. 107 North 2nd Road, Shihezi, Xinjiang, 832008, China.
| | - Chaoyang Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, 78 Hengzhigang Road, Guangzhou, 510095, China.
| |
Collapse
|
22
|
Sun C, Yang J, Cheng HB, Shen WX, Jiang ZQ, Wu MJ, Li L, Li WT, Chen TT, Rao XW, Zhou JR, Wu MH. 2-Hydroxy-3-methylanthraquinone inhibits lung carcinoma cells through modulation of IL-6-induced JAK2/STAT3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 61:152848. [PMID: 31035048 PMCID: PMC9618327 DOI: 10.1016/j.phymed.2019.152848] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/22/2019] [Accepted: 01/26/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND 2-hydroxy-3-methylanthraquinone (HMA), an anthraquinone monomer in traditional Chinese medicine Hedyotis diffusa, has been reported to inhibit the growth of several types of cancer, but its effect on lung cancer has not been adequately investigated. HYPOTHESIS/PURPOSE This study aimed to test the hypothesis that HMA inhibit the growth, migration, and invasion of lung cancer cells in part via downregulation of interleukin (IL)-6-induced JAK2/STAT3 pathway. METHODS Growth and apoptosis of lung cancer cells were quantitated by CCK-8 assay and Annexin V-FITC/PI flow cytometric analysis, respectively. Migration and invasion of A549 cells were determined by wound-healing assay and transwell invasion assay, respectively. The effect of HMA on cytokines expression in A549 cells was evaluated by the cytokine antibody array assay. Gene expression and protein levels of related molecular markers were quantitated by real time-PCR and Western blot analysis, respectively. RESULTS HMA significantly inhibited IL-6-stimulated growth and colony formation of A549 cells, increased the number of apoptotic cells, and inhibited invasion associated with downregulation of expression of IL-6-induced MMP-1, MMP-2, and MMP-9 genes. IL-6 increased the levels of tyrosine phosphorylation of JAK2 and STAT3 in A549 cells, which was reversed by HMA treatment. In addition, HMA reduced the expression of a series of inflammation-related cytokines in A549 cells supernatant, including IL-6, G-CSF, IL-6R, IL-8, MCP-1, RANTES, TNF-α. CONCLUSION These results suggest that HMA may inhibit the growth and invasion of lung cancer cells in part via downregulation of IL-6-induced JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Chao Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Institute of Oncology, the First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Translational Medicine Research Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Jing Yang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Institute of Oncology, the First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Hai-Bo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Translational Medicine Research Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Wei-Xing Shen
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Translational Medicine Research Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Ze-Qun Jiang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Institute of Oncology, the First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Ming-Jie Wu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Li Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Institute of Oncology, the First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Wen-Ting Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Institute of Oncology, the First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Ting-Ting Chen
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Translational Medicine Research Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Xi-Wu Rao
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Translational Medicine Research Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA.
| | - Mian-Hua Wu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Institute of Oncology, the First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
23
|
Kong C, Xie H, Gao Z, Shao M, Li H, Shi R, Cai L, Gao S, Sun T, Li C. Binding between Prion Protein and Aβ Oligomers Contributes to the Pathogenesis of Alzheimer's Disease. Virol Sin 2019; 34:475-488. [PMID: 31093882 DOI: 10.1007/s12250-019-00124-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 03/26/2019] [Indexed: 12/26/2022] Open
Abstract
A plethora of evidence suggests that protein misfolding and aggregation are underlying mechanisms of various neurodegenerative diseases, such as prion diseases and Alzheimer's disease (AD). Like prion diseases, AD has been considered as an infectious disease in the past decades as it shows strain specificity and transmission potential. Although it remains elusive how protein aggregation leads to AD, it is becoming clear that cellular prion protein (PrPC) plays an important role in AD pathogenesis. Here, we briefly reviewed AD pathogenesis and focused on recent progresses how PrPC contributed to AD development. In addition, we proposed a potential mechanism to explain why infectious agents, such as viruses, conduce AD pathogenesis. Microbe infections cause Aβ deposition and upregulation of PrPC, which lead to high affinity binding between Aβ oligomers and PrPC. The interaction between PrPC and Aβ oligomers in turn activates the Fyn signaling cascade, resulting in neuron death in the central nervous system (CNS). Thus, silencing PrPC expression may turn out be an effective treatment for PrPC dependent AD.
Collapse
Affiliation(s)
- Chang Kong
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Hao Xie
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhenxing Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ming Shao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Huan Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Run Shi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lili Cai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shanshan Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China. .,Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou, 510095, China.
| |
Collapse
|
24
|
Gao Z, Peng M, Chen L, Yang X, Li H, Shi R, Wu G, Cai L, Song Q, Li C. Prion Protein Protects Cancer Cells against Endoplasmic Reticulum Stress Induced Apoptosis. Virol Sin 2019; 34:222-234. [PMID: 31020572 PMCID: PMC6513834 DOI: 10.1007/s12250-019-00107-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/28/2019] [Indexed: 12/20/2022] Open
Abstract
Unfolded protein response (UPR) is an adaptive reaction for cells to reduce endoplasmic reticulum (ER) stress. In many types of cancers, such as lung cancer and pancreatic cancer, cancer cells may harness ER stress to facilitate their survival and growth. Prion protein (PrP) is a glycosylated cell surface protein that has been shown to be up-regulated in many cancer cells. Since PrP is a protein prone to misfolding, ER stress can result in under-glycosylated PrP, which in turn may activate ER stress. To assess whether ER stress leads to the production of under-glycosylated PrP and whether under-glycosylated PrP may contribute to ER stress thus leading to cancer cell apoptosis, we treated different cancer cells with brefeldin A (BFA), thapsigargin (Thps), and tunicamycin (TM). We found that although BFA, Thps, and TM treatment activated UPR, only ATF4 was consistently activated by these reagents, but not other branches of ER stress. However, the canonical PERK-eIF2α-ATF4 did not account for the observed activation of ATF4 in lung cancer cells. In addition, BFA, but neither Thps nor TM, significantly stimulated the expression of cytosolic PrP. Finally, we found that the levels of PrP contributed to anti-apoptosis activity of BFA-induced cancer cell death. Thus, the pathway of BFA-induced persistent ER stress may be targeted for lung and pancreatic cancer treatment.
Collapse
Affiliation(s)
- Zhenxing Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liang Chen
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaowen Yang
- Department of the First Abdominal Surgery, Jiangxi Tumor Hospital, Nanchang, 330029, China
| | - Huan Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Run Shi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Guiru Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lili Cai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qibin Song
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
25
|
Zhang C, Wang P, Li Y, Huang C, Ni W, Chen Y, Shi J, Chen G, Hu X, Ye M, Duan S, Wang K. Role of MicroRNAs in the Development of Hepatocellular Carcinoma in Nonalcoholic Fatty Liver Disease. Anat Rec (Hoboken) 2018; 302:193-200. [PMID: 30312023 DOI: 10.1002/ar.23954] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 02/24/2018] [Accepted: 03/09/2018] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent liver malignancy that can be developed from nonalcoholic fatty liver disease (NAFLD). Numerous pathophysiological alterations, including insulin resistance, specific cytokine release, oxidative stress, and mitochondrial damage, are involved in the transition of NAFLD to cirrhosis and HCC. MicroRNAs, as post-transcriptional modulators, play a critical role in the pathogenesis of NAFLD-related HCC by regulating lipid metabolism, glucose homeostasis, cell proliferation, apoptosis, migration, and differentiation. This review summarizes the current progress of microRNAs in the risk and prognosis of NAFLD-related HCC. Anat Rec, 302:193-200, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Ping Wang
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yongqiang Li
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Changxin Huang
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wei Ni
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yidan Chen
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Junping Shi
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Gongying Chen
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiangrong Hu
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Meng Ye
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Shiwei Duan
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Kaifeng Wang
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Salvesen Ø, Tatzelt J, Tranulis MA. The prion protein in neuroimmune crosstalk. Neurochem Int 2018; 130:104335. [PMID: 30448564 DOI: 10.1016/j.neuint.2018.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/04/2018] [Accepted: 11/14/2018] [Indexed: 01/11/2023]
Abstract
The cellular prion protein (PrPC) is a medium-sized glycoprotein, attached to the cell surface by a glycosylphosphatidylinositol anchor. PrPC is encoded by a single-copy gene, PRNP, which is abundantly expressed in the central nervous system and at lower levels in non-neuronal cells, including those of the immune system. Evidence from experimental knockout of PRNP in rodents, goats, and cattle and the occurrence of a nonsense mutation in goat that prevents synthesis of PrPC, have shown that the molecule is non-essential for life. Indeed, no easily recognizable phenotypes are associate with a lack of PrPC, except the potentially advantageous trait that animals without PrPC cannot develop prion disease. This is because, in prion diseases, PrPC converts to a pathogenic "scrapie" conformer, PrPSc, which aggregates and eventually induces neurodegeneration. In addition, endogenous neuronal PrPC serves as a toxic receptor to mediate prion-induced neurotoxicity. Thus, PrPC is an interesting target for treatment of prion diseases. Although loss of PrPC has no discernable effect, alteration of its normal physiological function can have very harmful consequences. It is therefore important to understand cellular processes involving PrPC, and research of this topic has advanced considerably in the past decade. Here, we summarize data that indicate the role of PrPC in modulating immune signaling, with emphasis on neuroimmune crosstalk both under basal conditions and during inflammatory stress.
Collapse
Affiliation(s)
- Øyvind Salvesen
- Faculty of Veterinary Medicine, Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Sandnes, Norway.
| | - Jörg Tatzelt
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany.
| | - Michael A Tranulis
- Faculty of Veterinary Medicine, Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| |
Collapse
|
27
|
Wang Z, Wang L, Wu X, Pan YL, Xie P, Pei G, Liang YN, Tang ZS, Liu L. Polysaccharide extracted from Portulacae Oleracea L. exerts protective effects against dextran sulfate sodium-induced colitis through inhibition of NF-κB. Am J Transl Res 2018; 10:2502-2510. [PMID: 30210688 PMCID: PMC6129545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/07/2018] [Indexed: 06/08/2023]
Abstract
Portulacae Oleracea L. (POL) is a traditional Chinese medicine and also an edible vegetable used to treat diarrhea in china for thousands years. Though the therapeutic effect has been proved in clinical trials, the concrete effective component and mechanisms remained elusive. Polysaccharide from POL has been extracted previously and the experiment suggested that POLP could diminish the weight loss and improve the health conditions of mice with DSS induced colitis. Hematoxylin & eosin staining revealed that POLP could improve the histopathological structure of the colon tissue. For the notably variation curve of TNF-α in control, colitis and treatment group, NF-κB was enrolled to investigate the molecular mechanisms of the protective effect of POLP. The protein expression level of NF-κBp65 in cytoplasm increased after POLP treatment of the induced colitis. However, the protein level of NF-κBp65 in the nucleus decreased after administration of POLP. The expression levels of IκBα and NF-κB related proteins Bcl-2 and survivin were also detected and the results suggested that POLP could inhibit the degradation of IκBα and decrease the protein levels of Bcl-2 and Survivin in colitis. It was concluded that POLP could improve the health condition of mice with DSS induced colitis and the mechanisms were closely related with NF-κB via inhibiting the degradation of IκBα.
Collapse
Affiliation(s)
- Zheng Wang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| | - Li Wang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| | - Xue Wu
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| | - Ya-Lei Pan
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| | - Pei Xie
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| | - Gang Pei
- School of Pharmacy, Hunan University of Chinese MedicineChangsha 410208, Hunan, China
| | - Yan-Ni Liang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| | - Zhi-Shu Tang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| | - Li Liu
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi Rheumatism and Tumor Center of TCM Engineering Technology Research, Shaanxi University of Chinese MedicineXianyang 712083, China
| |
Collapse
|
28
|
Linsenmeier L, Mohammadi B, Wetzel S, Puig B, Jackson WS, Hartmann A, Uchiyama K, Sakaguchi S, Endres K, Tatzelt J, Saftig P, Glatzel M, Altmeppen HC. Structural and mechanistic aspects influencing the ADAM10-mediated shedding of the prion protein. Mol Neurodegener 2018; 13:18. [PMID: 29625583 PMCID: PMC5889536 DOI: 10.1186/s13024-018-0248-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/21/2018] [Indexed: 11/10/2022] Open
Abstract
Background Proteolytic processing of the prion protein (PrPC) by endogenous proteases generates bioactive membrane-bound and soluble fragments which may help to explain the pleiotropic roles of this protein in the nervous system and in brain diseases. Shedding of almost full-length PrPC into the extracellular space by the metalloprotease ADAM10 is of peculiar relevance since soluble PrP stimulates axonal outgrowth and is protective in neurodegenerative conditions such as Alzheimer’s and prion disease. However, molecular determinates and mechanisms regulating the shedding of PrP are entirely unknown. Methods We produced an antibody recognizing the neo-epitope of shed PrP generated by ADAM10 in biological samples and used it to study structural and mechanistic aspects affecting the shedding. For this, we investigated genetically modified cellular and murine models by biochemical and morphological approaches. Results We show that the novel antibody specifically detects shed PrP in cell culture supernatants and murine brain. We demonstrate that ADAM10 is the exclusive sheddase of PrPC in the nervous system and reveal that the glycosylation state and type of membrane-anchorage of PrPC severely affect its shedding. Furthermore, we provide evidence that PrP shedding can be modulated by pharmacological inhibition and stimulation and present data suggesting that shedding is a relevant part of a compensatory network ensuring PrPC homeostasis of the cell. Conclusions With the new antibody, our study introduces a new tool to reliably investigate PrP-shedding. In addition, this study provides novel and important insight into the regulation of this cleavage event, which is likely to be relevant for diagnostic and therapeutic approaches even beyond neurodegeneration.
Collapse
Affiliation(s)
- Luise Linsenmeier
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | - Berta Puig
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Alexander Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Keiji Uchiyama
- Division of Molecular Neurobiology, Institute of Enzyme Research, Tokushima University, Tokushima, Japan
| | - Suehiro Sakaguchi
- Division of Molecular Neurobiology, Institute of Enzyme Research, Tokushima University, Tokushima, Japan
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Jörg Tatzelt
- Institute of Biochemistry and Pathobiochemistry, Ruhr University, Bochum, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Hermann C. Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
29
|
Wang SZ, Qin ZH. Anti-Inflammatory and Immune Regulatory Actions of Naja naja atra Venom. Toxins (Basel) 2018; 10:E100. [PMID: 29495566 PMCID: PMC5869388 DOI: 10.3390/toxins10030100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/23/2018] [Accepted: 02/24/2018] [Indexed: 02/06/2023] Open
Abstract
Naja naja atra venom (NNAV) is composed of various proteins, peptides, and enzymes with different biological and pharmacological functions. A number of previous studies have reported that NNAV exerts potent analgesic effects on various animal models of pain. The clinical studies using whole venom or active components have confirmed that NNAV is an effective and safe medicine for treatment of chronic pain. Furthermore, recent studies have demonstrated that NNAV has anti-inflammatory and immune regulatory actions in vitro and in vivo. In this review article, we summarize recent studies of NNAV and its components on inflammation and immunity. The main new findings in NNAV research show that it may enhance innate and humoral immune responses while suppressing T lymphocytes-mediated cellular immunity, thus suggesting that NNAV and its active components may have therapeutic values in the treatment of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China.
| |
Collapse
|
30
|
The function of the cellular prion protein in health and disease. Acta Neuropathol 2018; 135:159-178. [PMID: 29151170 DOI: 10.1007/s00401-017-1790-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Abstract
The essential role of the cellular prion protein (PrPC) in prion disorders such as Creutzfeldt-Jakob disease is well documented. Moreover, evidence is accumulating that PrPC may act as a receptor for protein aggregates and transduce neurotoxic signals in more common neurodegenerative disorders, such as Alzheimer's disease. Although the pathological roles of PrPC have been thoroughly characterized, a general consensus on its physiological function within the brain has not yet been established. Knockout studies in various organisms, ranging from zebrafish to mice, have implicated PrPC in a diverse range of nervous system-related activities that include a key role in the maintenance of peripheral nerve myelination as well as a general ability to protect against neurotoxic stimuli. Thus, the function of PrPC may be multifaceted, with different cell types taking advantage of unique aspects of its biology. Deciphering the cellular function(s) of PrPC and the consequences of its absence is not simply an academic curiosity, since lowering PrPC levels in the brain is predicted to be a powerful therapeutic strategy for the treatment of prion disease. In this review, we outline the various approaches that have been employed in an effort to uncover the physiological and pathological functions of PrPC. While these studies have revealed important clues about the biology of the prion protein, the precise reason for PrPC's existence remains enigmatic.
Collapse
|