1
|
Jiao P, Yang N, Jia Q, Fan B, Feng K, Yu J, Zhao S. A dual-reporter LDLR system integrating fluorescence and luminescence for understanding LDLR regulation and facilitating drug discovery. Front Mol Biosci 2025; 12:1552085. [PMID: 40182619 PMCID: PMC11966430 DOI: 10.3389/fmolb.2025.1552085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The low-density lipoprotein receptor (LDLR) is integral to cholesterol metabolism and cardiovascular health. Enhancing LDLR expression is a promising strategy for treating hyperlipidemia and reducing the risk of atherosclerosis. However, current LDLR reporter systems have limitations in detecting both transcriptional and translational regulation. To address this, we developed a novel dual-reporter LDLR system incorporating Enhanced Green Fluorescent Protein (EGFP) and Gaussia luciferase (Gluc) to enable precise monitoring of LDLR expression and function. Methods A CRISPR/Cas9-mediated knock-in strategy was used to integrate EGFP and Gluc upstream of the stop codon located in exon 18 of the LDLR gene in HEK293 cells. The dual-reporter system allows real-time visualization of LDLR expression via EGFP fluorescence and quantitative assessment through secreted Gluc activity. The system was validated using western blotting, immunofluorescence, and functional assays, including DiI-LDL uptake and drug response analyses with statins and PCSK9 inhibitors. Results The established LDLR-EGFP-Gluc knock-in cell line faithfully recapitulates endogenous LDLR expression and function. EGFP fluorescence accurately reflects LDLR expression dynamics, while Gluc activity provides a highly sensitive and quantitative readout. Functional assays confirmed that LDLR expression responds appropriately to statins and PCSK9 inhibitors. Additionally, screening for transcriptional regulators identified FOXP3 and CREB as novel modulators of LDLR expression, with CREB-mediated regulation involving the sterol regulatory element-binding protein 2 (SREBP2) pathway. Discussion This dual-reporter system enables complementary monitoring of LDLR dynamics, providing enhanced sensitivity, accuracy, and versatility for studying LDLR regulation and function, as well as facilitating drug discovery targeting hyperlipidemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Peng Jiao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Na Yang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Qianfeng Jia
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Baozhen Fan
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Ke Feng
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Jian Yu
- Department of Basic Medical Education, Yantai Nursing School, Yantai, Shandong, China
| | - Shengtian Zhao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
2
|
Spinsante C, Carducci F, Carotti E, Canapa A, Bizzaro D, Biscotti MA, Barucca M. A bioinformatic approach to characterize the vitellogenin receptor and the low density lipoprotein receptor superfamily in the newt Cynops orientalis. Sci Rep 2025; 15:3403. [PMID: 39870874 PMCID: PMC11772764 DOI: 10.1038/s41598-025-88011-6] [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: 07/16/2024] [Accepted: 01/23/2025] [Indexed: 01/29/2025] Open
Abstract
The Low Density Lipoprotein receptors (LDLRs) gene family includes 15 receptors: very low-density lipoprotein receptor (VLDLR), LDLR, Sorting-related receptor with A-type repeats (SORLA), and 12 LDL receptor-related proteins (LRPs): LRP1, LRP1B, LRP2, LRP3, LRP4, LRP5, LRP6, LRP8, LRP10, LRP11, LRP12, LRP13. Most of these are involved in the transduction of key signals during embryonic development and in the regulation of cholesterol homeostasis. In oviparous animals, the VLDL receptor is also known as VTGR since it facilitates the uptake of vitellogenin in ovary. In tetrapods, information concerning genes encoding these proteins is limited to a few taxa. Here, we report the characterization of VTGR in the amphibian Cynops orientalis. The secondary structure analyses and the expression profiles obtained from hepatic and gonadal tissues of C. orientalis supported the role of VTGR as vitellogenin oocyte membrane receptor in this species. Moreover, to get a holistic view of the evolutionary history of this gene superfamily, we extended our investigation to all 15 genes belonging to the LDLR superfamily analyzing through a phylogenetic analysis a total of 161 sequences belonging to 11 genera of vertebrates. The position of LRP8 in the tree and its expression findings in C. orientalis ovary allowed us to suggest that other proteins of the LDLR superfamily could act as receptors during vitellogenesis.
Collapse
Affiliation(s)
- Chiara Spinsante
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Federica Carducci
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Elisa Carotti
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Adriana Canapa
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Davide Bizzaro
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Maria Assunta Biscotti
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy.
| | - Marco Barucca
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| |
Collapse
|
3
|
Frey ZD, Price DA, Connors KA, Rush RE, Brown G, Sterling CE, Fatma F, Schwarz MM, Ganaie S, Cui X, Wills ZP, Leung DW, Amarasinghe GK, Hartman AL. Lrp1 facilitates infection of neurons by Jamestown Canyon virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622176. [PMID: 39574651 PMCID: PMC11580904 DOI: 10.1101/2024.11.06.622176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Jamestown Canyon virus (JCV) is a bunyavirus and arbovirus responsible for neuroinvasive disease in the United States. Little is known about JCV pathogenesis, and no host factors required for cellular infection have been identified. Recently, we identified low-density lipoprotein receptor related protein 1 (Lrp1) as a host entry factor for two other bunyaviruses Rift Valley fever virus (RVFV) and Oropouche virus (OROV). Here, we assessed the role of Lrp1 in mediating JCV cellular infection of neurons. Both neuronal and non-neuronal immortalized cell lines deficient for Lrp1 displayed reduction in infection with JCV, and early stages of infection such as binding and internalization were impacted by lack of Lrp1. In primary rat neurons, Lrp1 was highly expressed, and the neurons were highly permissive for JCV infection. Treatment of primary neurons with recombinant receptor-associated protein (RAP), a high affinity ligand for Lrp1, resulted in reduced infectivity with JCV. In addition, pretreatment of cells with RVFV Gn inhibited JCV infection, suggesting that the two viruses may share overlapping binding sites. These results provide compelling evidence that Lrp1 is an important cellular factor for efficient infection by JCV, and thus multiple bunyaviruses with varying clinical manifestations and tissue tropism are facilitated by the host cell Lrp1. Reliance of multiple bunyaviruses on Lrp1 makes it a promising target for pan-bunyaviral antivirals and therapeutics.
Collapse
Affiliation(s)
- Zachary D Frey
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - David A Price
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Kaleigh A Connors
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rachael E Rush
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Griffin Brown
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Cade E Sterling
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Farheen Fatma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Madeline M Schwarz
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Safder Ganaie
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Xiaoxia Cui
- Genome Engineering and Stem Cell Center (GEiC), Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Zachary P Wills
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Daisy W Leung
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Amy L Hartman
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
4
|
Cosset FL, Denolly S. Lipoprotein receptors: A little grease for enveloped viruses to open the lock? J Biol Chem 2024; 300:107849. [PMID: 39357828 PMCID: PMC11550601 DOI: 10.1016/j.jbc.2024.107849] [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: 08/07/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
Several studies recently highlighted the role of lipoprotein receptors in viral entry. These receptors are evolutionarily ancient proteins, key for the transport of lipids as well as other signaling molecules across the plasma membrane. Here, we discuss the different families of lipoprotein receptors and how they are hijacked by enveloped viruses to promote their entry into infected cells. While the usage of lipoprotein receptors was known for members of the Flaviviridae family and vesicular stomatitis virus, the last 4 years have seen the discovery that these receptors are used by many genetically unrelated viruses. We also emphasize how viral particles interact with these receptors and the possible targeting of these host factors as antiviral strategies.
Collapse
Affiliation(s)
- François-Loïc Cosset
- CIRI - Centre International de Recherche en Infectiologie, Université de Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308 ENS de Lyon, Lyon, France.
| | - Solène Denolly
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon1, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
5
|
Qu Z, Luo J, Li Z, Yang R, Zhao J, Chen X, Yu S, Shu H. Advancements in strategies for overcoming the blood-brain barrier to deliver brain-targeted drugs. Front Aging Neurosci 2024; 16:1353003. [PMID: 39253614 PMCID: PMC11381257 DOI: 10.3389/fnagi.2024.1353003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier is known to consist of a variety of cells and complex inter-cellular junctions that protect the vulnerable brain from neurotoxic compounds; however, it also complicates the pharmacological treatment of central nervous system disorders as most drugs are unable to penetrate the blood-brain barrier on the basis of their own structural properties. This dramatically diminished the therapeutic effect of the drug and compromised its biosafety. In response, a number of drugs are often delivered to brain lesions in invasive ways that bypass the obstruction of the blood-brain barrier, such as subdural administration, intrathecal administration, and convection-enhanced delivery. Nevertheless, these intrusive strategies introduce the risk of brain injury, limiting their clinical application. In recent years, the intensive development of nanomaterials science and the interdisciplinary convergence of medical engineering have brought light to the penetration of the blood-brain barrier for brain-targeted drugs. In this paper, we extensively discuss the limitations of the blood-brain barrier on drug delivery and non-invasive brain-targeted strategies such as nanomedicine and blood-brain barrier disruption. In the meantime, we analyze their strengths and limitations and provide outlooks on the further development of brain-targeted drug delivery systems.
Collapse
Affiliation(s)
- Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, China
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Juan Luo
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zheng Li
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Yang
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiaxi Zhao
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
6
|
Civeira F, Martín C, Cenarro A. APOE and familial hypercholesterolemia. Curr Opin Lipidol 2024; 35:195-199. [PMID: 38640077 DOI: 10.1097/mol.0000000000000937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
PURPOSE OF REVIEW Autosomal dominant hypercholesterolemia is a common cause of cardiovascular disease. In addition to the classic genes that cause hypercholesterolemia, LDLR, APOB and PCSK9 , a new locus has emerged as a candidate to be the cause of this hyperlipidemia, the p.(Leu167del) mutation in the APOE gene. RECENT FINDINGS Various studies have demonstrated the involvement of the p.(Leu167del) mutation in the APOE gene in hypercholesterolemia: Studies of family segregation, lipoprotein composition by ultracentrifugation and proteomic techniques, and functional studies of VLDL-carrying p.(Leu167del) internalization with cell cultures have demonstrated the role of this mutation in the cause of hypercholesterolemia. The phenotype of individuals carrying the p.(Leu167del) in APOE is indistinguishable from familial hypercholesterolemia individuals with mutations in the classic genes. However, a better response to lipid-lowering treatment has been demonstrated in these APOE mutation carrier individuals. SUMMARY Therefore, APOE gene should be considered a candidate locus along with LDLR, APOB , and PCSK9 to be investigated in the genetic diagnosis of familial hypercholesterolemia.
Collapse
Affiliation(s)
- Fernando Civeira
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV
- Universidad de Zaragoza, Zaragoza
| | - César Martín
- Biofisika Institute (UPV/EHU, CSIC), University of the Basque Country, Leioa
- Department of Biochemistry and Molecular Biology, UPV/EHU, University of the Basque Country, Bilbao
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| |
Collapse
|
7
|
André S, Verteneuil S, Ris L, Kahvecioglu ZC, Nonclercq D, De Winter J, Vander Elst L, Laurent S, Muller RN, Burtea C. Modulation of Cytosolic Phospholipase A2 as a Potential Therapeutic Strategy for Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1395-1426. [PMID: 38225969 PMCID: PMC10789292 DOI: 10.3233/adr-230075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/17/2023] [Indexed: 01/17/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder lacking any curative treatment up to now. Indeed, actual medication given to the patients alleviates only symptoms. The cytosolic phospholipase A2 (cPLA2-IVA) appears as a pivotal player situated at the center of pathological pathways leading to AD and its inhibition could be a promising therapeutic approach. Objective A cPLA2-IVA inhibiting peptide was identified in the present work, aiming to develop an original therapeutic strategy. Methods We targeted the cPLA2-IVA using the phage display technology. The hit peptide PLP25 was first validated in vitro (arachidonic acid dosage [AA], cPLA2-IVA cellular translocation) before being tested in vivo. We evaluated spatial memory using the Barnes maze, amyloid deposits by MRI and immunohistochemistry (IHC), and other important biomarkers such as the cPLA2-IVA itself, the NMDA receptor, AβPP and tau by IHC after i.v. injection in APP/PS1 mice. Results Showing a high affinity for the C2 domain of this enzyme, the peptide PLP25 exhibited an inhibitory effect on cPLA2-IVA activity by blocking its binding to its substrate, resulting in a decreased release of AA. Coupled to a vector peptide (LRPep2) in order to optimize brain access, we showed an improvement of cognitive abilities of APP/PS1 mice, which also exhibited a decreased number of amyloid plaques, a restored expression of cPLA2-IVA, and a favorable effect on NMDA receptor expression and tau protein phosphorylation. Conclusions cPLA2-IVA inhibition through PLP25 peptide could be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Séverine André
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sébastien Verteneuil
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Laurence Ris
- Department of Neurosciences, University of Mons, Research Institute for Health Science and Technologies, Mons, Belgium
| | - Zehra-Cagla Kahvecioglu
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | | | - Julien De Winter
- Organic Synthesis and Mass Spectrometry Laboratory (SMOs), University of Mons-UMONS, Mons, Belgium
| | - Luce Vander Elst
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Robert N. Muller
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Carmen Burtea
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| |
Collapse
|
8
|
Lane AN, Nash PD, Ellsworth SA, Nystrom GS, Rokyta DR. The arylsulfatase- and phospholipase-rich venom of the plutoniumid centipede Theatops posticus. Toxicon 2023; 233:107231. [PMID: 37517595 DOI: 10.1016/j.toxicon.2023.107231] [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: 06/17/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
Research on centipede venoms has led to the discovery of a diverse array of novel proteins and peptides, including those with homology to previously discovered toxin families (e.g., phospholipase A2s and pM12a metalloproteases) and novel toxin families not previously detected in venoms (e.g., β-pore forming toxins and scoloptoxins). Most of this research has focused on centipedes in the order Scolopendromorpha, particularly those in the families Scolopendridae, Cryptopidae, and Scolopocryptopidae. To generate the first high-throughput venom characterization for a centipede in the scolopendromorph family Plutoniumidae, we performed venom-gland transcriptomics and venom proteomics on two Theatops posticus. We identified a total of 64 venom toxins, 60 of which were detected in both the venom-gland transcriptome and venom proteome and four of which were only detected transcriptomically. We detected a single highly abundant arylsulfatase B (ARSB) toxin, the first ARSB toxin identified from centipede venoms. As ARSBs have been detected in other venomous species (e.g., scorpions), ARSBs in T. posticus highlights a new case of convergent evolution across venoms. Theatops posticus venom also contained a much higher abundance and diversity of phospholipase A2 toxins compared to other characterized centipede venoms. Conversely, we detected other common centipedes toxins, such as CAPs and scoloptoxins, at relatively low abundances and diversities. Our observation of a diverse set of toxins from T. posticus venom, including those from novel toxin families, emphasizes the importance of studying unexplored centipede taxonomic groups and the continued potential of centipede venoms for novel toxin discovery and unraveling the molecular mechanisms underlying trait evolution.
Collapse
Affiliation(s)
- Aaliyah N Lane
- Department of Biological Science, Florida State University, Tallahassee, FL, 32306, USA
| | - Pauline D Nash
- Department of Biological Science, Florida State University, Tallahassee, FL, 32306, USA
| | - Schyler A Ellsworth
- Department of Biological Science, Florida State University, Tallahassee, FL, 32306, USA
| | - Gunnar S Nystrom
- Department of Biological Science, Florida State University, Tallahassee, FL, 32306, USA
| | - Darin R Rokyta
- Department of Biological Science, Florida State University, Tallahassee, FL, 32306, USA.
| |
Collapse
|
9
|
Zulijani A, Milardović A, Kovač Z, Perić B, Jakovac H. Megalin Expression in Primary Oral Squamous Cell Carcinoma Is Associated with the Presence of Lymph Node Metastases, Vascular Invasion, and Lower Overall Survival. Curr Issues Mol Biol 2023; 45:2757-2766. [PMID: 37185704 PMCID: PMC10136934 DOI: 10.3390/cimb45040180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Megalin (LRP2) is a rapidly recycling multiligand endocytic receptor primarily expressed in polarized epithelial cells. Although megalin might be involved in tumor growth and invasiveness through several mechanisms, its role has been understudied in the field of molecular oncology so far. The present study aimed to evaluate the impact of megalin expression in oral squamous cell carcinoma (OSCC) on disease progression. Megalin expression was evaluated immunohistochemically in 63 OSCC specimens. Data obtained were retrospectively compared with patient clinicopathological features and their survival. The proportion of megalin-expressing cells in the primary OSCC tissue was significantly associated with metastatic spreading to lymph nodes, vascular invasion and lower overall survival rate. Results obtained by the study suggest that megalin can be considered as a novel molecule involved in OSCC pathogenesis, but also useful as a potential biomarker for cancer progression.
Collapse
|
10
|
Babio L, Damsteegt EL, Lokman PM. Lipoprotein receptors in ovary of eel, Anguilla australis: molecular characterisation of putative vitellogenin receptors. FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:117-137. [PMID: 36648592 PMCID: PMC9935665 DOI: 10.1007/s10695-023-01169-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
Lipoprotein receptors, including low-density lipoprotein receptor (LDLr) relatives (Lrs) and LDLr-related proteins (Lrps), belong to the LDLr supergene family and participate in diverse physiological functions. In this study, novel sequences of lr and lrp genes expressed in the ovary of the short-finned eel, Anguilla australis, during early gonadal development are presented. The genes encoding the LDLr-like, Lrp1-like, Lrp1b-like, Lrp3, Lrp4-like, Lrp5-like, Lrp6, Lrp10, Lrp11, Lrp12-like, and Lr11-like proteins were found and identified by sequence and structure analysis, in addition to phylogenetic analysis. Genes encoding proteins previously implicated in follicle development and vitellogenin (Vtg) uptake in oviparous vertebrates were also identified, i.e. lr8 (including lr8 + and lr8- variants) and lrp13; their identification was reinforced by conserved synteny with orthologues in other teleost fish. Compared to other lr/lrp genes, the genes encoding Lr8 + , Lr8-, and Lrp13 were highly expressed in ovary during early development, decreasing as oocyte development advanced when induced by hypophysation. Furthermore, lr8 + , lr8-, and lrp13 were dominantly expressed in the ovary when compared with 17 other tissues. Finally, this study successfully detected the expression of both lr8 variants, which showed different expression patterns to those reported in other oviparous vertebrates and provided the first characterisation of Lrp13 in Anguilla sp. We propose that lr8 + , lr8-, and lrp13 encode putative Vtg receptors in anguillid eels.
Collapse
Affiliation(s)
- Lucila Babio
- Department of Zoology, University of Otago, 340 Great King Street, P.O. Box 56, Dunedin, Otago 9054 New Zealand
| | - Erin L. Damsteegt
- Department of Zoology, University of Otago, 340 Great King Street, P.O. Box 56, Dunedin, Otago 9054 New Zealand
| | - P. Mark Lokman
- Department of Zoology, University of Otago, 340 Great King Street, P.O. Box 56, Dunedin, Otago 9054 New Zealand
| |
Collapse
|
11
|
LDLR, LRP1, and Megalin redundantly participate in the uptake of Clostridium novyi alpha-toxin. Commun Biol 2022; 5:906. [PMID: 36064583 PMCID: PMC9445046 DOI: 10.1038/s42003-022-03873-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 08/23/2022] [Indexed: 11/30/2022] Open
Abstract
Clostridium novyi alpha-toxin (Tcnα) is a potent exotoxin that induces severe symptoms including gas gangrene, myositis, necrotic hepatitis, and sepsis. Tcnα binds to sulfated glycosaminoglycans (sGAG) for cell-surface attachment and utilizes low-density lipoprotein receptor (LDLR) for rapid entry. However, it was also shown that Tcnα may use alternative entry receptors other than LDLR. Here, we define that LRP1 and Megalin can also facilitate the cellular entry of Tcnα by employing reconstitutive LDLR family proteins. LDLR, LRP1, and Megalin recognize Tcnα via their ligand-binding domains (also known as LDL receptor type A repeats). Notably, LDLR and LRP1 have contrasting expression levels in many different cells, thus the dominant entry receptor for Tcnα could be cell-type dependent. These findings together increase our knowledge of the Tcnα actions and further help to understand the pathogenesis of C. novyi infection-associated diseases. Clostridium novyi alpha-toxin (Tcnα) also uses LRP1 and Megalin as cellular entry receptors besides LDLR, and this might be a response to cell-type dependent receptor availability for the exotoxin.
Collapse
|
12
|
Passarella D, Ciampi S, Di Liberto V, Zuccarini M, Ronci M, Medoro A, Foderà E, Frinchi M, Mignogna D, Russo C, Porcile C. Low-Density Lipoprotein Receptor-Related Protein 8 at the Crossroad between Cancer and Neurodegeneration. Int J Mol Sci 2022; 23:ijms23168921. [PMID: 36012187 PMCID: PMC9408729 DOI: 10.3390/ijms23168921] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
The low-density-lipoprotein receptors represent a family of pleiotropic cell surface receptors involved in lipid homeostasis, cell migration, proliferation and differentiation. The family shares common structural features but also has significant differences mainly due to tissue-specific interactors and to peculiar proteolytic processing. Among the receptors in the family, recent studies place low-density lipoprotein receptor-related protein 8 (LRP8) at the center of both neurodegenerative and cancer-related pathways. From one side, its overexpression has been highlighted in many types of cancer including breast, gastric, prostate, lung and melanoma; from the other side, LRP8 has a potential role in neurodegeneration as apolipoprotein E (ApoE) and reelin receptor, which are, respectively, the major risk factor for developing Alzheimer’s disease (AD) and the main driver of neuronal migration, and as a γ-secretase substrate, the main enzyme responsible for amyloid formation in AD. The present review analyzes the contributions of LDL receptors, specifically of LRP8, in both cancer and neurodegeneration, pointing out that depending on various interactions and peculiar processing, the receptor can contribute to both proliferative and neurodegenerative processes.
Collapse
Affiliation(s)
- Daniela Passarella
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Silvia Ciampi
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, 90133 Palermo, Italy
| | - Mariachiara Zuccarini
- Department of Medical Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Alessandro Medoro
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Emanuele Foderà
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Monica Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, 90133 Palermo, Italy
| | - Donatella Mignogna
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| | - Claudio Russo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
- Correspondence: ; Tel.: +39-0874404897
| | - Carola Porcile
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
| |
Collapse
|
13
|
The Role of ApoE Serum Levels and ApoE Gene Polymorphisms in Patients with Laryngeal Squamous Cell Carcinoma. Biomolecules 2022; 12:biom12081013. [PMID: 35892323 PMCID: PMC9331506 DOI: 10.3390/biom12081013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Recent studies have revealed that the inflammatory ApoE effect may play a significant role in various cancer development. However, this effect has still not been analyzed in patients with laryngeal squamous cell carcinoma (LSCC). In the present study, we evaluated two single nucleotide polymorphisms (SNPs) of ApoE (rs7412 and rs429358) and determined their associations with LSCC development and the LSCC patients’ five-year survival rate. Additionally, we analyzed serum ApoE levels using an enzyme-linked immunosorbent assay. A total of 602 subjects (291 histologically verified LSCC patients and 311 healthy controls) were involved in this study. The genotyping was carried out using the real-time PCR. We revealed that ApoE ε3/ε3 was associated with a 1.7-fold higher probability of developing LSCC (p = 0.001), with 1.7-fold increased odds of developing LSCC without metastasis to the lymph nodes (p = 0.002) and with a 2.0-fold increased odds of developing well-differentiated LSCC (p = 0.008), as well as 1.6-fold increased odds of developing poorly differentiated LSCC development (p = 0.012). The ApoE ε2/ε4 and ε3/ε4 genotypes were associated with a 2.9-fold and 1.5-fold decrease in the likelihood of developing LSCC (p = 0.042; p = 0.037, respectively). ApoE ε3/ε4 was found associated with a 2.4-fold decreased likelihood of developing well-differentiated LSCC (p = 0.013). Conclusion: ApoE ε2/ε4 and ε3/ε4 were found to play a protective role in LSCC development, while ApoE ε3/ε3 may have a risk position in LSCC development.
Collapse
|
14
|
Wan H, Zhong J, Zhang Z, Zou P, Wang Y. Comparative Transcriptome Reveals the Potential Modulation Mechanisms of Spfoxl-2 Affecting Ovarian Development of Scylla paramamosain. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:125-135. [PMID: 35107659 DOI: 10.1007/s10126-022-10091-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
Previously, we reported the identification, tissue distribution and confirmed the roles of Spfoxl-2 in regulating vitellogenin (vtg) expression in Scylla paramamosain. Here, we primally analyzed its potential target genes in the ovary with RNAi and RNA-Seq technology. By comparing the transcriptome data of two groups (ovaries that injected with EGFP and Foxl-2 siRNA, respectively), we found 645 DEGs (differentially expressed genes), including several conserved crucial genes involved in ovarian development, such as vtg, vitellogenin receptor (vtgR), adenylate cyclase (AC), cyclinB, and cell division cycle 2 (cdc2). In addition, these DEGs were also enriched in pathways related to ovary development, including relaxin signaling pathway, ovarian steroidogenesis, and progesterone-mediated oocyte maturation. Moreover, several genes were selected for qRT-PCR to validate the accuracy of the bioinformatic result. To the best of our knowledge, the current study was the first report about foxl-2 function through comparative transcriptome analysis in crustacean species, which identified not only relevant genes and pathways involved in ovarian development of S. paramamosain, but also provided new insights into the regulatory mechanisms of foxl-2 at the molecular level in crustacean.
Collapse
Affiliation(s)
- Haifu Wan
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- Fujian Engineering Research Center of Aquatic Breeding and Healthy Aquaculture, Xiamen, 361021, China
| | - Jinying Zhong
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- Fujian Engineering Research Center of Aquatic Breeding and Healthy Aquaculture, Xiamen, 361021, China
| | - Ziping Zhang
- College of Marine Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Pengfei Zou
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- Fujian Engineering Research Center of Aquatic Breeding and Healthy Aquaculture, Xiamen, 361021, China
| | - Yilei Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China.
- Fujian Engineering Research Center of Aquatic Breeding and Healthy Aquaculture, Xiamen, 361021, China.
| |
Collapse
|
15
|
Su H, Ren W, Zhang D. Research progress on exosomal proteins as diagnostic markers of gastric cancer (review article). Clin Exp Med 2022; 23:203-218. [DOI: 10.1007/s10238-022-00793-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 01/04/2022] [Indexed: 12/20/2022]
Abstract
AbstractGastric cancer (GC) is one of the most common types of tumors and the most common cause of cancer mortality worldwide. The diagnosis of GC is critical to its prevention and treatment. Available tumor markers are the crucial step for GC diagnosis. Recent studies have shown that proteins in exosomes are potential diagnostic and prognostic markers for GC. Exosomes, secreted by cells, are cup-shaped with a diameter of 30–150 nm under the electron microscope. They are also surrounded by lipid bilayers and are widely found in various body fluids. Exosomes contain proteins, lipids and nucleic acid. The examination of exosomal proteins has the advantages of quickness, easy sampling, and low pain and cost, as compared with the routine inspection method of GC, which may lead to marked developments in GC diagnosis. This article summarized the exosomal proteins with a diagnostic and prognostic potential in GC, as well as exosomal proteins involved in GC progression.
Collapse
|
16
|
Lin J, Zheng Z, Liu J, Yang G, Leng L, Wang H, Qiu G, Wu Z. LRP5-Mediated Lipid Uptake Modulates Osteogenic Differentiation of Bone Marrow Mesenchymal Stromal Cells. Front Cell Dev Biol 2021; 9:766815. [PMID: 34796178 PMCID: PMC8593169 DOI: 10.3389/fcell.2021.766815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
Nutritional microenvironment determines the specification of progenitor cells, and lipid availability was found to modulate osteogenesis in skeletal progenitors. Here, we investigated the implications of lipid scarcity in the osteogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs) and the role of low-density lipoprotein receptor-related protein 5 (LRP5), a co-receptor transducing canonical Wnt/beta-catenin signals, in BMSC lipid uptake during osteogenesis. The osteogenic differentiation of murine BMSCs was suppressed by lipid scarcity and partially rescued by additional fatty acid treatment with oleate. The enhancement of osteogenesis by oleate was found to be dosage-dependent, along with the enhanced activation of beta-catenin and Wnt target genes. Conditional knockout (CKO) of Lrp5 gene in murine mesenchymal lineage using Lrp5fl/fl;Prrx1-cre mice led to decreased bone quality and altered fat distribution in vivo. After Lrp5 ablation using adenoviral Cre-recombinase, the accumulation of lipid droplets in BMSC cytoplasm was significantly reduced, and the osteogenesis of BMSCs was suppressed. Moreover, the impaired osteogenesis due to either lipid scarcity or Lrp5 ablation could be rescued by recombinant Wnt3a protein, indicating that the osteogenesis induced by Wnt/beta-catenin signaling was independent of LRP5-mediated lipid uptake. In conclusion, lipid scarcity suppresses BMSC osteogenic differentiation. LRP5 plays a role in the uptake of lipids in BMSCs and therefore mediates osteogenic specification.
Collapse
Affiliation(s)
- Jiachen Lin
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhifa Zheng
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jieying Liu
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guihua Yang
- Harmony Technology Co., Ltd., Beijing, China
| | - Ling Leng
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihong Wu
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Zhang YM. Orosomucoid-like protein 3, rhinovirus and asthma. World J Crit Care Med 2021; 10:170-182. [PMID: 34616654 PMCID: PMC8462028 DOI: 10.5492/wjccm.v10.i5.170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/16/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
The genetic variants of orosomucoid-like protein 3 (ORMDL3) gene are associated with highly significant increases in the number of human rhinovirus (HRV)-induced wheezing episodes in children. Recent investigations have been focused on the mechanisms of ORMDL3 in rhinovirus infection for asthma and asthma exacerbations. ORMDL3 not only regulates major human rhinovirus receptor intercellular adhesion molecule 1 expression, but also plays pivotal roles in viral infection through metabolisms of ceramide and sphingosine-1-phosphate, endoplasmic reticulum (ER) stress, ER-Golgi interface and glycolysis. Research on the roles of ORMDL3 in HRV infection will lead us to identify new biomarkers and novel therapeutic targets in childhood asthma and viral induced asthma exacerbations.
Collapse
Affiliation(s)
- You-Ming Zhang
- Section of Genomic and Environmental Medicine, National Heart and Lung Institute, Molecular Genetics Group, Division of Respiratory Sciences, Imperial College London, London SW3 6LY, United Kingdom
| |
Collapse
|
18
|
Zulijani A, Dekanić A, Ćabov T, Jakovac H. Metallothioneins and Megalin Expression Profiling in Premalignant and Malignant Oral Squamous Epithelial Lesions. Cancers (Basel) 2021; 13:4530. [PMID: 34572758 PMCID: PMC8464971 DOI: 10.3390/cancers13184530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/28/2022] Open
Abstract
This study aimed to assess the relationship and possible interactions between metallothioneins (MTs) and megalin (LRP-2) in different grades of oral squamous cell carcinoma (OSCC) and premalignant lesions of the oral mucosa (oral leukoplakia and oral lichen planus). The study included archived samples of 114 patients and control subjects. Protein expression was examined by immunohistochemistry and immunofluorescence, and staining quantification was performed by ImageJ software. Protein interaction in cancer tissue was tested and visualized by proximity ligation assay. Mann-Whitney and Kruskal-Wallis tests were used to determine the significance of differences between each group, whereas Pearson correlation coefficient was performed to test correlation. Expression of both proteins differed significantly between each group showing the same pattern of gradual increasing from oral lichen planus to poorly differentiated OSCC. Moreover, MTs and megalin were found to co-express and interact in cancer tissue, and their expression positively correlated within the overall study group. Findings of prominent nuclear and chromosomal megalin expression suggest that it undergoes regulated intramembrane proteolysis upon MTs binding, indicating its ability to directly affect gene expression and cellular division in cancer tissue. The data obtained point to the onco-driving potential of MTs-megalin interaction.
Collapse
Affiliation(s)
- Ana Zulijani
- Department of Oral Surgery, Clinical Hospital Center Rijeka, Krešimirova ul. 40, 51000 Rijeka, Croatia;
| | - Andrea Dekanić
- Department of Pathology, Clinical Hospital Center Rijeka, Krešimirova ul. 42, 51000 Rijeka, Croatia;
| | - Tomislav Ćabov
- Faculty of Dental Medicine, University of Rijeka, Krešimirova ul. 40, 51000 Rijeka, Croatia
| | - Hrvoje Jakovac
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Ul. Braće Branchetta 20, 51000 Rijeka, Croatia
| |
Collapse
|
19
|
Li Y, Luo G. Human low-density lipoprotein receptor plays an important role in hepatitis B virus infection. PLoS Pathog 2021; 17:e1009722. [PMID: 34293069 PMCID: PMC8345860 DOI: 10.1371/journal.ppat.1009722] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 08/06/2021] [Accepted: 06/17/2021] [Indexed: 01/05/2023] Open
Abstract
Hepatitis B virus (HBV) chronically infects more than 240 million people worldwide, resulting in chronic hepatitis, cirrhosis, and hepatocellular carcinoma. HBV vaccine is effective to prevent new HBV infection but does not offer therapeutic benefit to hepatitis B patients. Neither are current antiviral drugs curative of chronic hepatitis B. A more thorough understanding of HBV infection and replication holds a great promise for identification of novel antiviral drugs and design of optimal strategies towards the ultimate elimination of chronic hepatitis B. Recently, we have developed a robust HBV cell culture system and discovered that human apolipoprotein E (apoE) is enriched on the HBV envelope and promotes HBV infection and production. In the present study, we have determined the role of the low-density lipoprotein receptor (LDLR) in HBV infection. A LDLR-blocking monoclonal antibody potently inhibited HBV infection in HepG2 cells expressing the sodium taurocholate cotransporting polypeptide (NTCP) as well as in primary human hepatocytes. More importantly, small interfering RNAs (siRNAs)-mediated knockdown of LDLR expression and the CRISPR/Cas9-induced knockout of the LDLR gene markedly reduced HBV infection. A recombinant LDLR protein could block heparin-mediated apoE pulldown, suggesting that LDLR may act as an HBV cell attachment receptor via binding to the HBV-associated apoE. Collectively, these findings demonstrate that LDLR plays an important role in HBV infection probably by serving as a virus attachment receptor. Requirement of multiple cell surface receptors and co-receptors for efficient virus infection is exemplified by human immunodeficient virus (HIV) and hepatitis C virus (HCV). In the case of HBV, expression of the NTCP receptor alone in human and murine hepatocytes converted HBV susceptibility albeit at low levels. Recent identification of the glypican 5 (GPC5) and epidermal growth factor receptor (EGFR) as HBV infection-promoting factors suggests that efficient HBV infection requires multiple cell surface molecules as virus attachment and post-attachment receptors. Here, we provide substantial evidence demonstrating that another cell surface receptor LDLR plays an important role in HBV infection. Downregulation of LDLR expression significantly lowered HBV infection, whereas its upregulation promoted HBV infection. The levels of LDLR expression correlated with HBV cell attachment, suggesting that it serves as an HBV cell attachment receptor. The inhibition of heparin-mediated apoE pulldown by a purified LDLR suggested that LDLR promotes HBV infection probably through its binding to HBV-associated apoE. It is warranted to further determine whether other LDLR family members also play a role in HBV infection.
Collapse
Affiliation(s)
- Yingying Li
- Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States of America
- Department of Microbiology, Peking University Health Science Center School of Basic Medical Sciences, Beijing, China
| | - Guangxiang Luo
- Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
20
|
Guo S, Chen Y, Liu J, Zhang X, Liu Z, Zhou Z, Wei W. Low-density lipoprotein receptor-related protein 1 is a CROPs-associated receptor for Clostridioides difficile toxin B. SCIENCE CHINA-LIFE SCIENCES 2021; 65:107-118. [PMID: 34279819 DOI: 10.1007/s11427-021-1943-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/28/2021] [Indexed: 12/23/2022]
Abstract
As the leading cause of worldwide hospital-acquired infection, Clostridioides difficile (C. difficile) infection has caused heavy economic and hospitalized burden, while its pathogenesis is not fully understood. Toxin B (TcdB) is one of the major virulent factors of C. difficile. Recently, CSPG4 and FZD2 were reported to be the receptors that mediate TcdB cellular entry. However, genetic ablation of genes encoding these receptors failed to completely block TcdB entry, implicating the existence of alternative receptor(s) for this toxin. Here, by employing the CRISPR-Cas9 screen in CSPG4-deficient HeLa cells, we identified LDL receptor-related protein-1 (LRP1) as a novel receptor for TcdB. Knockout of LRP1 in both CSPG4-deficient HeLa cells and colonic epithelium Caco2 cells conferred cells with increased TcdB resistance, while LRP1 overexpression sensitized cells to TcdB at a low concentration. Co-immunoprecipitation assay showed that LRP1 interacts with full-length TcdB. Moreover, CROPs domain, which is dispensable for TcdB's interaction with CSPG4 and FZD2, is sufficient for binding to LRP1. As such, our study provided evidence for a novel mechanism of TcdB entry and suggested potential therapeutic targets for treating C. diff.
Collapse
Affiliation(s)
- Shengjie Guo
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yiou Chen
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Jingze Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xinyi Zhang
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zhiheng Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zhuo Zhou
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China.
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
21
|
Abstract
Large clostridial toxins (LCTs) are a family of bacterial exotoxins that infiltrate and destroy target cells. Members of the LCT family include Clostridioides difficile toxins TcdA and TcdB, Paeniclostridium sordellii toxins TcsL and TcsH, Clostridium novyi toxin TcnA, and Clostridium perfringens toxin TpeL. Since the 19th century, LCT-secreting bacteria have been isolated from the blood, organs, and wounds of diseased individuals, and LCTs have been implicated as the primary virulence factors in a variety of infections, including C. difficile infection and some cases of wound-associated gas gangrene. Clostridia express and secrete LCTs in response to various physiological signals. LCTs invade host cells by binding specific cell surface receptors, ultimately leading to internalization into acidified vesicles. Acidic pH promotes conformational changes within LCTs, which culminates in translocation of the N-terminal glycosyltransferase and cysteine protease domain across the endosomal membrane and into the cytosol, leading first to cytopathic effects and later to cytotoxic effects. The focus of this review is on the role of LCTs in infection and disease, the mechanism of LCT intoxication, with emphasis on recent structural work and toxin subtyping analysis, and the genomic discovery and characterization of LCT homologues. We provide a comprehensive review of these topics and offer our perspective on emerging questions and future research directions for this enigmatic family of toxins.
Collapse
|
22
|
Ouyang Q, Meng Y, Zhou W, Tong J, Cheng Z, Zhu Q. New advances in brain-targeting nano-drug delivery systems for Alzheimer's disease. J Drug Target 2021; 30:61-81. [PMID: 33983096 DOI: 10.1080/1061186x.2021.1927055] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide and its incidence is increasing due to the ageing population. Currently, the main limitations of AD treatment are low blood-brain barrier permeability, severe off-target of drugs, and immune abnormality. In this review, four hypotheses for Alzheimer's pathogenesis and three challenges for Alzheimer's drug delivery are discussed. In addition, this article summarises the different strategies of brain targeting nano-drug delivery systems (NDDSs) developed in the last 10 years. These strategies include receptor-mediated (transferrin receptor, low-density lipoprotein receptor-related protein, lactoferrin receptor, etc.), adsorption-mediated (cationic, alkaline polypeptide, cell-penetrating peptides, etc.), and transporter-mediated (P-gp, GLUT1, etc.). Moreover, it provides insights into novel strategies used in AD, such as exosomes, virus-like particles, and cell membrane coating particles. Hence, this review will help researchers to understand the current progress in the field of NDDSs for the central nervous system and find new directions for AD therapy.HighlightsCharacteristics and challenges based on the pathogenesis of AD were discussed.Recent advances in novel brain-targeting NDDSs for AD over the past 10 years were summarised.
Collapse
Affiliation(s)
- Qin Ouyang
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Yingcai Meng
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Jianbin Tong
- Department of Anaesthesiology, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China.,Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zeneng Cheng
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| | - Qubo Zhu
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
Wang C, Zou Y, Pan C, Shao L, Ding Z, Zhang Y, Ye J, Li P, Ren Y, Zhu C. Prognostic significance of chemokines CCL11 and CCL5 modulated by low-density lipoprotein cholesterol in colon cancer patients with normal body mass index. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:202. [PMID: 33708829 PMCID: PMC7940920 DOI: 10.21037/atm-20-1604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Many studies have shown an elevated level of cholesterol in colon tumors as compared to normal tissue. Obesity and high low-density lipoprotein cholesterol (LDL-C) are known risk factors for colon cancer. However, the role of LDL-C in colon cancer patients with normal body mass index (BMI) remains elusive. Methods Levels of serum cholesterol and oxysterols were quantified by ultra-performance liquid chromatography-tandem mass spectrometer (UPLC-MS/MS) from 129 individuals with normal BMI, including 32 with solitary polyp, 36 with multiple polyps, and 31 with adenocarcinoma as well as 32 healthy controls. In vitro, colon cancer cells were treated with LDL-C and assayed for chemokines via RNA-Seq and mitochondrial morphology via transmission electron microscopy and immunofluorescence. Additionally, correlation analysis was performed between LDL-C-induced chemokines and the overall survival of colon cancer patients from the Cancer Genome Atlas (TCGA), the Genotype-Tissue Expression (GTEx), and the Human Protein Atlas (HPA) database. Results The serum cholesterol level was significantly higher in colon adenocarcinoma patients with normal BMI than that in healthy controls (P<0.001). LDL-C potentiated colon cancer cell invasion and resistance to glucose-deprivation in vitro via chemokine-mediated signaling, mainly upregulation of CC chemokine ligand (CCL) 5 and downregulation of CCL 11. By analyzing the RNA expression data of colorectal cancer from TCGA, GTEx, and HPA, we demonstrated that the CCL5 level in colorectal adenocarcinoma tissues was significantly increased relative to adjacent normal tissues (P=0.01) while the CCL11 level was decreased (P=0.01). Both increased CCL5 and decreased CCL11 showed a negative correlation with the 5-year overall survival in tumor node metastasis (TNM) stage II colon cancer patients (P=0.0032, 0.026 for CCL5 and CCL11, respectively). Conclusions Our study supports the idea that LDL-C regulates the expression of CCL5 and CCL11 chemokines, which may have predictive values for survival in colon cancer patients with normal BMI, especially for patients in TNM stage II.
Collapse
Affiliation(s)
- Caihua Wang
- Department of Gastroenterology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Zou
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chi Pan
- Department of Surgical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liming Shao
- Department of Gastroenterology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zonghui Ding
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Yunzhu Zhang
- Department of Gastroenterology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Ye
- Department of Gastroenterology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peiwei Li
- Department of Gastroenterology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuezhong Ren
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunpeng Zhu
- Department of Gastroenterology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
24
|
Chen Q, Zhang Y, Zhao Q. Expression analysis of immune-associated genes in hemocytes of mud crab Scylla paramamosain under low salinity challenge. FISH & SHELLFISH IMMUNOLOGY 2020; 107:16-25. [PMID: 32947031 DOI: 10.1016/j.fsi.2020.09.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 06/11/2023]
Abstract
To gain knowledge on the immune response in Scylla paramamosain under low salinity challenge, S. paramamosain we investigated digital gene expression (DEG) in S. paramamosain hemocytes using the deep-sequencing platform Illumina Hiseq XTen. A total of 97,257 high quality unigenes with mean length 786.59 bp were found to be regulated by low salinity challenge, among which 93 unigenes were significantly up regulated, and 71 were significantly down regulated. Functional categorization and pathways analysis of differentially expressed genes revealed that immune signaling pathway including cAMP and cGMP signaling pathway were affected in low salinity stress. Cellular immunity-related genes including low-density lipoprotein receptor-related protein 6 (LRP6) and xanthine dehydrogenase (XDH) were down-regulated, indicating phagocytosis and oxygen dependent mechanism of phagocyte were suppressed in low salinity stress; Humoral immunity-related genes serine proteases and serpins 3 were up- and down-regulated, respectively, suggest that the proPO system was influenced by low salinity significantly; Moreover, processes related to immune response including carbohydrate metabolism, protein synthesis and lipid transport were found differentially regulated, implying the integrity of the immune response in low salinity stress. This study gained comprehensive insights on the immune mechanism of S. paramamosain at low salinity stress at the molecular level. The findings provide a theoretical basis for understanding immune mechanisms of S. paramamosain under low salinity stress, and technical reference for evaluating physiological adaptation in fresh water environment.
Collapse
Affiliation(s)
- Qinsheng Chen
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Yan Zhang
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Qun Zhao
- Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan Aquaculture Breeding Engineering Research Center, College of Marine Sciences, Hainan University, Haikou, Hainan, 570228, China.
| |
Collapse
|
25
|
Low-density lipoprotein receptor (LDLR) regulates NLRP3-mediated neuronal pyroptosis following cerebral ischemia/reperfusion injury. J Neuroinflammation 2020; 17:330. [PMID: 33153475 PMCID: PMC7643474 DOI: 10.1186/s12974-020-01988-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 10/07/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Inflammatory response has been recognized as a pivotal pathophysiological process during cerebral ischemic stroke. NLRP3 inflammasome, involved in the regulation of inflammatory cascade, can simultaneously lead to GSDMD-executed pyroptosis in cerebral ischemia. Low-density lipoprotein receptor (LDLR), responsible for cholesterol uptake, was noted to exert potential anti-inflammatory bioactivities. Nevertheless, the role of LDLR in neuroinflammation mobilized by cerebral ischemia/reperfusion (I/R) has not been investigated. METHODS Ischemic stroke mice model was accomplished by middle cerebral artery occlusion. Oxygen-glucose deprivation was employed after primary cortical neuron was extracted and cultured. A pharmacological inhibitor of NLRP3 (CY-09) was administered to suppress NLPR3 activation. Histological and biochemical analysis were performed to assess the neuronal death both in vitro and in vivo. In addition, neurological deficits and behavioral deterioration were evaluated in mice. RESULTS The expression of LDLR was downregulated following cerebral I/R injury. Genetic knockout of Ldlr enhanced caspase-1-dependent cleavage of GSDMD and resulted in severe neuronal pyroptosis. LDLR deficiency contributed to excessive NLRP3-mediated maturation and release of IL-1β and IL-18 under in vitro and in vivo ischemic conditions. These influences ultimately led to aggravated neurological deficits and long-term cognitive dysfunction. Blockade of NLRP3 substantially retarded neuronal pyroptosis in Ldlr-/- mice and cultured Ldlr-/- neuron after experimental stroke. CONCLUSIONS These results demonstrated that LDLR modulates NLRP3-mediated neuronal pyroptosis and neuroinflammation following ischemic stroke. Our findings characterize a novel role for LDLR as a potential therapeutic target in neuroinflammatory responses to acute cerebral ischemic injury.
Collapse
|
26
|
Al‐Bari AA, Al Mamun A. Current advances in regulation of bone homeostasis. FASEB Bioadv 2020; 2:668-679. [PMID: 33205007 PMCID: PMC7655096 DOI: 10.1096/fba.2020-00058] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
Bone homeostasis is securely controlled by the dynamic well-balanced actions among osteoclasts, osteoblasts and osteocytes. Osteoclasts are large multinucleated cells that degrade bone matrix and involve in the bone remodelling in conjunction with other bone cells, osteoblasts and osteocytes, the completely matured form of osteoblasts. Disruption of this controlling balance among these cells or any disparity in bone remodelling caused by a higher rate of resorption by osteoclasts over construction of bone by osteoblasts results in a reduction of bone matrix including bone mineral density (BMD) and bone marrow cells (BMCs). The dominating effect of osteoclasts results in advanced risk of bone crack and joint destruction in several diseases including osteoporosis and rheumatoid arthritis (RA). However, the boosted osteoblastic activity produces osteosclerotic phenotype and weakened its action primes to osteomalacia or rickets. On the other hand, senescent osteocytes predominately progress the senescence associated secretory phenotype (SASP) and may contribute to age related bone loss. Here, we discuss an advanced level work on newly identified cellular mechanisms controlling the remodelling of bone and crosstalk among bone cells as these relate to the therapeutic targeting of the skeleton.
Collapse
Affiliation(s)
| | - Abdullah Al Mamun
- Department of Genetic Engineering and BiotechnologyShahjalal University of Science and TechnologySylhetBangladesh
| |
Collapse
|
27
|
Jakovac H, Stašić N, Krašević M, Jonjić N, Radošević-Stašić B. Expression profiles of metallothionein-I/II and megalin/LRP-2 in uterine cervical squamous lesions. Virchows Arch 2020; 478:735-746. [PMID: 33084977 PMCID: PMC7990851 DOI: 10.1007/s00428-020-02947-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/18/2020] [Accepted: 10/09/2020] [Indexed: 12/29/2022]
Abstract
Metallothioneins (MTs) are phylogenetically old cysteine-rich proteins, which are implicated in a variety of physiological and pathological processes. Their growth-regulating, anti-apoptotic and anti-inflammatory functions have been attributed not only to intracellular free radical scavenging and to zinc and copper regulation but also to the ability of secreted MT to bind on surface lipoprotein receptor-megalin/LRP2, which enables the endocytosis of MT-I/II and a wide range of other functionally distinct ligands. In the present study, we analysed the expression pattern of both proteins in 55 cases of premalignant transformation of cervical squamous cells, i.e. in low- and high-grade squamous intraepithelial lesion (LSIL and HSIL). The data showed that in LSIL (cervical intraepithelial neoplasia CIN1; N = 25) MTs were present only in basal and parabasal cells and that megalin was only weakly expressed. In HSIL (CIN2; N = 15 and CIN 3/carcinoma in situ; N = 15), however, overexpression and co-localization of MT with megalin were found in the entire hyperplastic epithelium. Moreover, megalin immunoreactivity appeared on the glandular epithelium and vascular endothelium, as well as on lymphatic cells in stroma. Besides, multiple megalin-positive cells expressed phosphorylated Akt1, implying that MT- and/or megalin-dependent prosurvival signal transduction pathways might contribute to the development of severe cervical dysplasia. The data emphasize the diagnostic power of combined MT/megalin analysis in pre-cancer screening.
Collapse
Affiliation(s)
- Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Nikola Stašić
- Teaching Institute of Public Health, Primorsko-goranska County, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - Maja Krašević
- Department of Pathology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - Nives Jonjić
- Department of Pathology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - Biserka Radošević-Stašić
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| |
Collapse
|
28
|
Xue Y, Huang S, Huang J, Li S, Zhang C, Zhou X. Identification of Apolipoprotein E as a Potential Diagnostic Biomarker of Nasopharyngeal Carcinoma. Cancer Manag Res 2020; 12:8943-8950. [PMID: 33061590 PMCID: PMC7522425 DOI: 10.2147/cmar.s239479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 07/20/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose Apo-E, a secreted protein, is closely related to the migration and invasion of tumor cells. In this study, we aimed to analyze the expression of Apo-E in nasopharyngeal carcinoma (NPC) patients and cell lines, as well as its effects on NPC cell behavior. Patients and Methods Our study included 35 patients with NPC from Zhongnan Hospital. Expression levels of Apo-E in patients with NPC were examined by quantitative reverse transcription-polymerase chain reaction, Western blot, and immunohistochemical (IHC) staining. Receiver operating characteristic (ROC) curves were analyzed using the SPSS 22 software to estimate the sensitivity and specificity of the Apo-E protein in diagnosing NPC. Additionally, the level of Apo-E in NPC cell lines (NP69, 6–10B, and 5–8F) was investigated by Western blotting and IHC. Results Levels of Apo-E were higher in NPC patients than in controls. Moreover, ROC analysis revealed that increased Apo-E in the serum of NPC patients may act as a potential biomarker for NPC diagnosis (Area under the curve 0.917). Furthermore, similar results were also identified in NPC cancer cell lines. RNA interference technology was used to overexpress the endogenous Apo-E in the NPC cell line 6–10B. Wound healing and transwell assays indicated that the overexpression of Apo-E increased the number of cell colonies and migration ability, respectively. Conclusion In this study, we found that Apo-E was elevated in NPC patients and may act as a potential biomarker for NPC diagnosis. In addition, Apo-E was upregulated in NPC cell lines and promoted cell growth, migration, and invasion.
Collapse
Affiliation(s)
- Ying Xue
- The Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Shuo Huang
- The Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Jing Huang
- The Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Shuang Li
- The Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Cen Zhang
- The Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| | - Xuhong Zhou
- The Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, Hubei, People's Republic of China
| |
Collapse
|
29
|
Li Y, Rao Y, Zhu H, Jiang B, Zhu M. USP16 Regulates the Stability and Function of LDL receptor by Deubiquitination. Int Heart J 2020; 61:1034-1040. [PMID: 32999190 DOI: 10.1536/ihj.20-043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Low-density lipoprotein (LDL) particles are known to be atherogenic agents in coronary artery diseases. They adjust to other electronegative forms and can be the subject for the enhancement of inflammatory events in vessel subendothelial spaces. The LDL uptake is related to the membrane scavenger receptors, including LDL receptor (LDLR). The LDLR expression is closely associated with LDL uptake and occurrence of diseases, such as atherosclerotic cardiovascular diseases. Our findings identified USP16 as a novel regulator of LDLR due to its ability to prevent ubiquitylation-dependent LDLR degradation, further promoting the uptake of LDL. The enhancement of USP16-mediated deubiquitination andthe suppressive degradation of the LDLR cause the presentation of a potential strategy to increase LDL cholesterol clearance.
Collapse
Affiliation(s)
- Yongsheng Li
- Department of Cardiovascular Internal Medicine, The Fifth Hospital of Xiamen
| | - Yanbiao Rao
- Department of Cardiovascular Internal Medicine, The Fifth Hospital of Xiamen
| | - Hongtao Zhu
- Department of Cardiovascular Internal Medicine, The Fifth Hospital of Xiamen
| | - Bingyuan Jiang
- Department of Cardiovascular Internal Medicine, The Fifth Hospital of Xiamen
| | - Maoshu Zhu
- Central Laboratory, The Fifth Hospital of Xiamen
| |
Collapse
|
30
|
Kaźmierczak Z, Szostak-Paluch K, Przybyło M, Langner M, Witkiewicz W, Jędruchniewicz N, Dąbrowska K. Endocytosis in cellular uptake of drug delivery vectors: Molecular aspects in drug development. Bioorg Med Chem 2020; 28:115556. [PMID: 32828419 DOI: 10.1016/j.bmc.2020.115556] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022]
Abstract
Drug delivery vectors are widely applied to increase drug efficacy while reducing the side effects and potential toxicity of a drug. They allow for patient-tailored therapy, dose titration, and therapeutic drug monitoring. A major part of drug delivery systems makes use of large nanocarriers: liposomes or virus-like particles (VLPs). These systems allow for a relatively large amount of cargo with good stability of vectors, and they offer multiple options for targeting vectors in vivo. Here we discuss endocytic pathways that are available for drug delivery by large nanocarriers. We focus on molecular aspects of the process, including an overview of potential molecular targets for studies of drug delivery vectors and for future solutions allowing targeted drug delivery.
Collapse
Affiliation(s)
- Zuzanna Kaźmierczak
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Kamila Szostak-Paluch
- Research and Development Center, Regional Specialized Hospital, Wrocław, Poland; Wrocław University of Science and Technology, Faculty of Fundamental Technical Problems, Department of Biomedical Engineering, Wrocław, Poland
| | - Magdalena Przybyło
- Wrocław University of Science and Technology, Faculty of Fundamental Technical Problems, Department of Biomedical Engineering, Wrocław, Poland; Lipid Systems sp z o.o., Wrocław, Poland
| | - Marek Langner
- Wrocław University of Science and Technology, Faculty of Fundamental Technical Problems, Department of Biomedical Engineering, Wrocław, Poland; Lipid Systems sp z o.o., Wrocław, Poland
| | - Wojciech Witkiewicz
- Research and Development Center, Regional Specialized Hospital, Wrocław, Poland
| | | | - Krystyna Dąbrowska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland; Research and Development Center, Regional Specialized Hospital, Wrocław, Poland.
| |
Collapse
|
31
|
Ruan Y, Wong NK, Zhang X, Zhu C, Wu X, Ren C, Luo P, Jiang X, Ji J, Wu X, Hu C, Chen T. Vitellogenin Receptor (VgR) Mediates Oocyte Maturation and Ovarian Development in the Pacific White Shrimp ( Litopenaeus vannamei). Front Physiol 2020; 11:485. [PMID: 32499719 PMCID: PMC7243368 DOI: 10.3389/fphys.2020.00485] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/20/2020] [Indexed: 11/13/2022] Open
Abstract
Oocyte maturation and ovarian development are sequentially coordinated events critical to reproduction. In the ovaries of adult oviparous animals such as birds, bony fish, insects, and crustaceans, vitellogenin receptor (VgR) is a plasma membrane receptor that specifically mediates vitellogenin (Vg) transport into oocytes. Accumulation of Vg drives sexual maturation of the female crustaceans by acting as a pivotal regulator of nutritional accumulation within oocytes, a process known as vitellogenesis. However, the mechanisms by which VgR mediates vitellogenesis are still not fully understood. In this study, we first identified a unique VgR (Lv-VgR) and characterized its genomic organization and protein structural domains in Litopenaeus vannamei, a predominant cultured shrimp species worldwide. This newly identified Lv-VgR phylogenetically forms a group with VgRs from other crustacean species within the arthropod cluster. Duplicated LBD/EGFD regions are found exclusively among arthropod VgRs but not in paralogs from vertebrates and nematodes. In terms of expression patterns, Lv-VgR transcripts are specifically expressed in ovaries of female shrimps, which increases progressively during ovarian development, and rapidly declines toward embryonic development. The cellular and subcellular locations were For analyzed by in situ hybridization and immunofluorescence, respectively. The Lv-VgR mRNA was found to be expressed in the oocytes of ovaries, and Lv-VgR protein was found to localize in the cell membrane of maturing oocytes while accumulation of the ligand Vg protein assumed an even cytoplasmic distribution. Silencing of VgR transcript expression by RNAi was effective for stunting ovarian development. This present study has thus provided new insights into the regulatory roles of VgR in crustacean ovarian development.
Collapse
Affiliation(s)
- Yao Ruan
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Nai-Kei Wong
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Xin Zhang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunhua Zhu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Xiaofen Wu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunhua Ren
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,Institution of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, Guangzhou, China
| | - Peng Luo
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,Institution of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, Guangzhou, China
| | - Xiao Jiang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,Institution of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, Guangzhou, China
| | - Jiatai Ji
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Haimao Investment Co., Ltd., Zhanjiang, China
| | - Xugan Wu
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Chaoqun Hu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,Institution of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, Guangzhou, China
| | - Ting Chen
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology (LMB), Guangdong Provincial Key Laboratory of Applied Marine Biology (LAMB), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China.,Institution of South China Sea Ecology and Environmental Engineering (ISEE), Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
32
|
Ju S, Park S, Lim L, Choi DH, Song H. Low density lipoprotein receptor-related protein 1 regulates cardiac hypertrophy induced by pressure overload. Int J Cardiol 2019; 299:235-242. [PMID: 31350035 DOI: 10.1016/j.ijcard.2019.07.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Cardiac hypertrophy is associated with functional changes in cardiomyocytes, which often results in heart failure. The low-density lipoprotein receptor-related protein 1 (LRP1) is a large multifunctional endocytic receptor involved in many physiological and pathological processes. However, its function in the development of cardiac hypertrophy remains largely unclear. METHODS Adenoviral constructs were used for either overexpression or silencing of LRP1 in both in vitro and in vivo experiments. Cardiac function was measured using the Millar catheter. RESULTS LRP1 expression was upregulated in both transverse aortic constriction (TAC)-induced hypertrophic myocardium and catecholamine (phenylephrine (PE) and norepinephrine (NE))- and angiotensin II (AngII)-induced hypertrophic cardiomyocytes. In addition, cell surface area, protein/DNA ratio, and the mRNA levels of hypertrophic markers were significantly increased in LRP1-overexpressing cardiomyocytes without catecholamine stimulation. Conversely, LRP1 inhibition by LRP1-specific siRNA or a specific ligand-binding antagonist (RAP) significantly rescued hypertrophic effects in PE, NE, or AngII-induced cardiomyocytes. LRP1 overexpression induced PKCα, then activated ERK, resulting in cardiac hypertrophy with the downregulation of SERCA2a and calcium accumulation, which was successfully restored in both LRP1-silenced cardiomyocytes and TAC-induced hearts. CONCLUSIONS LRP1 regulates cardiac hypertrophy via the PKCα-ERK dependent signaling pathway resulting in the alteration of intracellular calcium levels, demonstrating that LRP1 might be a potential therapeutic target for cardiac hypertrophy.
Collapse
Affiliation(s)
- Sujin Ju
- Department of Medical of Sciences, Chosun University Graduate School, Gwangju 61452, Republic of Korea
| | - Seulki Park
- Department of Medical of Sciences, Chosun University Graduate School, Gwangju 61452, Republic of Korea
| | - Leejin Lim
- Department of Medical of Sciences, Chosun University Graduate School, Gwangju 61452, Republic of Korea; Cancer Mutation Research Center, Chosun University, Gwangju 61452, Republic of Korea
| | - Dong-Hyun Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju 61452, Republic of Korea
| | - Heesang Song
- Department of Medical of Sciences, Chosun University Graduate School, Gwangju 61452, Republic of Korea; Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju 61452, Republic of Korea.
| |
Collapse
|
33
|
Huang HC, Lin H, Huang MC. Lactoferrin promotes hair growth in mice and increases dermal papilla cell proliferation through Erk/Akt and Wnt signaling pathways. Arch Dermatol Res 2019; 311:411-420. [PMID: 31006055 PMCID: PMC6546667 DOI: 10.1007/s00403-019-01920-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/04/2019] [Accepted: 04/13/2019] [Indexed: 12/11/2022]
Abstract
Hair loss affects men and women of all ages. Dermal papilla (DP) plays a crucial role in regulating the growth and cycling of hair follicles. Lactoferrin (LF) exhibits a wide range of biological functions, including antimicrobial activity and growth regulation. However, its effect on DP and its role in hair growth remain unknown. In this study, we found that bovine LF (bLF) promoted the proliferation of DP cells and enhanced the phosphorylation of Erk and Akt. The bLF-mediated proliferation was significantly blocked by the Erk phosphorylation inhibitor PD98059 or the Akt phosphorylation inhibitor LY294002. Moreover, biotin-labeled bLF could bind to DP cells, and the binding was independent of lipoprotein receptor-related protein 1, a known LF receptor. Importantly, bLF stimulated hair growth in both young and aged mice. Moreover, we also found that bLF significantly induced the expression of Wnt signaling-related proteins, including Wnt3a, Wnt7a, Lef1, and β-catenin. The bLF-mediated DP cell proliferation could be significantly reversed by the Wnt pathway inhibitor XAV939. Our findings suggest that bLF promotes hair growth in mice and stimulates proliferation of DP cells through Erk/Akt and Wnt signaling pathways. This study highlights a great potential of the use of bLF in developing drugs to treat hair loss.
Collapse
Affiliation(s)
| | - Hsuan Lin
- Renorigin Innovation Institute Co. Ltd., Taipei, Taiwan
| | - Min-Chuan Huang
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec. 1, Ren'ai Road, Taipei, 100, Taiwan.
| |
Collapse
|
34
|
Basnet S, Palmenberg AC, Gern JE. Rhinoviruses and Their Receptors. Chest 2019; 155:1018-1025. [PMID: 30659817 DOI: 10.1016/j.chest.2018.12.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/14/2018] [Accepted: 12/28/2018] [Indexed: 01/14/2023] Open
Abstract
Human rhinoviruses (RVs) are picornaviruses that can cause a variety of upper and lower respiratory tract illnesses, including the common cold, bronchitis, pneumonia, and exacerbations of chronic respiratory diseases such as asthma. There are currently > 160 known types of RVs classified into three species (A, B, and C) that use three different cellular membrane glycoproteins expressed in the respiratory epithelium to enter the host cell. These viral receptors are intercellular adhesion molecule 1 (used by the majority of RV-A and all RV-B types), low-density lipoprotein receptor family members (used by 12 RV-A types), and cadherin-related family member 3 (CDHR3; used by RV-C). RV-A and RV-B interactions with intercellular adhesion molecule 1 and low-density lipoprotein receptor glycoproteins are well defined and their cellular functions have been described, whereas the mechanisms of the RV-C interaction with CDHR3 and its cellular functions are being studied. A single nucleotide polymorphism (rs6967330) in CDHR3 increases cell surface expression of this protein and, as a result, also promotes RV-C infections and illnesses. There are currently no approved vaccines or antiviral therapies available to treat or prevent RV infections, which is a major unmet medical need. Understanding interactions between RV and cellular receptors could lead to new insights into the pathogenesis of respiratory illnesses as well as lead to new approaches to control respiratory illnesses caused by RV infections.
Collapse
Affiliation(s)
- Sarmila Basnet
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI.
| | - Ann C Palmenberg
- Institute of Molecular Virology, University of Wisconsin-Madison, Madison, WI
| | - James E Gern
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
35
|
Guevara J, Romo J, Hernandez E, Guevara NV. Identification of Receptor Ligands in Apo B100 Reveals Potential Functional Domains. Protein J 2019; 37:548-571. [PMID: 30259240 DOI: 10.1007/s10930-018-9792-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
LDL, VLDL and other members of the low-density lipoparticles (LLPs) enter cells through a large family of receptors. The actual receptor ligand(s) in apolipoprotein B100, one of the main proteins of LLP, remain(s) unknown. The objective of this study was to identify true receptor ligand(s) in apo B100, a molecule of 4563 residues. Apo B100 contains 33 analogues of Cardin-Weintraub arginine/lysine-based receptor ligand motifs and shares key lysine motifs and sequence similarity with the LDL receptor-associated protein, MESD, and heat shock proteins. Eleven FITC-labeled synthetic peptides of 21-42 residues, with at least one ligand, were tested for binding and internalization using HeLa cells. All peptides bind but display different binding capacities and patterns. Peptides B0013, B0582, B2366, and B2932 mediate endocytosis and appear in distinct sites in the cytoplasm. B0708 and B3181 bind and remain on the cell surface as aggregates/clusters. Peptides B3119 (Site A) and B3347 (Site B), the putative ligands, showed low binding and no cell entry capacity. Apo B100 regions in this study share similarities with related proteins of known function including chaperone proteins and Apo BEC stimulating protein, and not directly related proteins, e.g., the DNA-binding domain of interferon regulatory factors, MSX2-interacting protein, and snake venom Zinc metalloproteinase-disintegrin-like proteins.
Collapse
Affiliation(s)
- Juan Guevara
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Jamie Romo
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Ernesto Hernandez
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Natalia Valentinova Guevara
- Biophysics Research Laboratory, Department of Physics and Astronomy, The University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA.
| |
Collapse
|
36
|
Deep sequencing and miRNA profiles in alcohol-induced neuroinflammation and the TLR4 response in mice cerebral cortex. Sci Rep 2018; 8:15913. [PMID: 30374194 PMCID: PMC6206094 DOI: 10.1038/s41598-018-34277-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
Alcohol abuse can induce brain injury and neurodegeneration, and recent evidence shows the participation of immune receptors toll-like in the neuroinflammation and brain damage. We evaluated the role of miRNAs as potential modulators of the neuroinflammation associated with alcohol abuse and the influence of the TLR4 response. Using mice cerebral cortex and next-generation sequencing (NGS), we identified miRNAs that were differentially expressed in the chronic alcohol-treated versus untreated WT or TLR4-KO mice. We observed a differentially expression of miR-183 Cluster (C) (miR-96/-182/-183), miR-200a and miR-200b, which were down-regulated, while mirR-125b was up-regulated in alcohol-treated WT versus (vs.) untreated mice. These miRNAs modulate targets genes related to the voltage-gated sodium channel, neuron hyperexcitability (Nav1.3, Trpv1, Smad3 and PP1-γ), as well as genes associated with innate immune TLR4 signaling response (Il1r1, Mapk14, Sirt1, Lrp6 and Bdnf). Functional enrichment of the miR-183C and miR-200a/b family target genes, revealed neuroinflammatory pathways networks involved in TLR4 signaling and alcohol abuse. The changes in the neuroinflammatory targets genes associated with alcohol abuse were mostly abolished in the TLR4-KO mice. Our results show the relationship between alcohol intake and miRNAs expression and open up new therapeutically targets to prevent deleterious effects of alcohol on the brain.
Collapse
|
37
|
Song JS, Seong HS, Choi BH, Lee CW, Hwang NH, Lim D, Lee JH, Kim JS, Kim JD, Park YS, Choi JW, Kim JB. Genome-wide analysis of Hanwoo and Chikso populations using the BovineSNP50 genotyping array. Genes Genomics 2018; 40:1373-1382. [DOI: 10.1007/s13258-018-0733-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 08/22/2018] [Indexed: 02/04/2023]
|
38
|
Jakovac H, Grubić Kezele T, Radošević-Stašić B. Expression Profiles of Metallothionein I/II and Megalin in Cuprizone Model of De- and Remyelination. Neuroscience 2018; 388:69-86. [PMID: 30025861 DOI: 10.1016/j.neuroscience.2018.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/20/2018] [Accepted: 07/05/2018] [Indexed: 11/15/2022]
Abstract
Copper chelator cuprizone (CPZ) is neurotoxicant, which selectively disrupts oligodendroglial respiratory chain, leading to oxidative stress and subsequent apoptosis. Demyelination is, however, followed by spontaneous remyelination owing to the activation of intrinsic CNS repair mechanisms. To explore the participation of metallothioneins (MTs) in these processes, in this study we analyzed the expression profiles of MT-I/II and their receptor megalin (low-density lipoprotein receptor related protein-2) in the brain of mice subjected to different protocols of CPZ feeding. Experiments were performed in female C57BL/6 mice fed with 0.25% CPZ during 1, 3 and 5 weeks. They were sacrificed immediately after feeding with CPZ or 2 weeks after the withdrawal of CPZ. The data showed that CPZ-induced demyelination was followed by high astrogliosis and enhanced expression of MTs and megalin in white (corpus callosum and internal capsule) and gray matter of the brain (cortex, hippocampus, and cerebellum). Moreover, in numerous cortical neurons and progenitor cells the signs of MT/megalin interactions and Akt1 phosphorylation was found supporting the hypothesis that MTs secreted from the astrocytes might directly affect the neuronal differentiation and survival. Furthermore, in mice treated with CPZ for 5 weeks the prominent MTs and megalin immunoreactivities were found on several neural stem cells and oligodendrocyte progenitors in subgranular zone of dentate gyrus and subventricular zone of lateral ventricles pointing to high modulatory effect of MTs on adult neuro- and oligodendrogenesis. The data show that MT I/II perform important cytoprotective and growth-regulating functions in remyelinating processes activated after toxic demyelinating insults.
Collapse
Affiliation(s)
- Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 RIJEKA, Croatia
| | - Tanja Grubić Kezele
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 RIJEKA, Croatia
| | - Biserka Radošević-Stašić
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 RIJEKA, Croatia.
| |
Collapse
|
39
|
Capillary morphogenesis gene 2 maintains gastric cancer stem-like cell phenotype by activating a Wnt/β-catenin pathway. Oncogene 2018; 37:3953-3966. [PMID: 29662192 PMCID: PMC6053357 DOI: 10.1038/s41388-018-0226-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/29/2017] [Accepted: 02/23/2018] [Indexed: 02/07/2023]
Abstract
A growing body of evidence shows that the development and progression of gastric cancer (GC) is mainly associated to the presence of gastric cancer stem-like cells (GCSLCs). However, it is unclear how GCSLC population is maintained. This study aimed to explore the role of capillary morphogenesis gene 2 (CMG2) in GCSLC maintenance and the relevance to GC progression. We found that CMG2 was highly expressed in GC tissues and the expression levels were associated with the invasion depth and lymph node metastasis of GC, and inversely correlated with the survival of GC patients. Sorted CMG2High GC cells preferentially clustered in CD44High stem-like cell population, which expressed high levels of stemness-related genes with increased capabilities of self-renewal and tumorigenicity. Depletion of CMG2 gene resulted in reduction of GCSLC population with attenuated stemness and decrease of invasive and metastatic capabilities with subdued epithelial–mesenchymal transition phenotype in GC cells. Mechanistically, CMG2 interacted with LRP6 in GCSLCs to activate a Wnt/β-catenin pathway. Thus, our results demonstrate that CMG2 promotes GC progression by maintaining GCSLCs and can serve as a new prognostic indicator and a target for human GC therapy.
Collapse
|
40
|
David M, Lécorché P, Masse M, Faucon A, Abouzid K, Gaudin N, Varini K, Gassiot F, Ferracci G, Jacquot G, Vlieghe P, Khrestchatisky M. Identification and characterization of highly versatile peptide-vectors that bind non-competitively to the low-density lipoprotein receptor for in vivo targeting and delivery of small molecules and protein cargos. PLoS One 2018; 13:e0191052. [PMID: 29485998 PMCID: PMC5828360 DOI: 10.1371/journal.pone.0191052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/27/2017] [Indexed: 01/09/2023] Open
Abstract
Insufficient membrane penetration of drugs, in particular biotherapeutics and/or low target specificity remain a major drawback in their efficacy. We propose here the rational characterization and optimization of peptides to be developed as vectors that target cells expressing specific receptors involved in endocytosis or transcytosis. Among receptors involved in receptor-mediated transport is the LDL receptor. Screening complex phage-displayed peptide libraries on the human LDLR (hLDLR) stably expressed in cell lines led to the characterization of a family of cyclic and linear peptides that specifically bind the hLDLR. The VH411 lead cyclic peptide allowed endocytosis of payloads such as the S-Tag peptide or antibodies into cells expressing the hLDLR. Size reduction and chemical optimization of this lead peptide-vector led to improved receptor affinity. The optimized peptide-vectors were successfully conjugated to cargos of different nature and size including small organic molecules, siRNAs, peptides or a protein moiety such as an Fc fragment. We show that in all cases, the peptide-vectors retain their binding affinity to the hLDLR and potential for endocytosis. Following i.v. administration in wild type or ldlr-/- mice, an Fc fragment chemically conjugated or fused in C-terminal to peptide-vectors showed significant biodistribution in LDLR-enriched organs. We have thus developed highly versatile peptide-vectors endowed with good affinity for the LDLR as a target receptor. These peptide-vectors have the potential to be further developed for efficient transport of therapeutic or imaging agents into cells -including pathological cells-or organs that express the LDLR.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Karine Varini
- VECT-HORUS SAS, Marseille, France
- Aix Marseille Univ, CNRS, NICN, Marseille, France
| | | | - Géraldine Ferracci
- Aix Marseille Univ, CNRS, CRN2M, Marseille, France
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | | | | | - Michel Khrestchatisky
- Aix Marseille Univ, CNRS, NICN, Marseille, France
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| |
Collapse
|
41
|
Skronska-Wasek W, Gosens R, Königshoff M, Baarsma HA. WNT receptor signalling in lung physiology and pathology. Pharmacol Ther 2018; 187:150-166. [PMID: 29458107 DOI: 10.1016/j.pharmthera.2018.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The WNT signalling cascades have emerged as critical regulators of a wide variety of biological aspects involved in lung development as well as in physiological and pathophysiological processes in the adult lung. WNTs (secreted glycoproteins) interact with various transmembrane receptors and co-receptors to activate signalling pathways that regulate transcriptional as well as non-transcriptional responses within cells. In physiological conditions, the majority of WNT receptors and co-receptors can be detected in the adult lung. However, dysregulation of WNT signalling pathways contributes to the development and progression of chronic lung pathologies, including idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma and lung cancer. The interaction between a WNT and the (co-)receptor(s) present at the cell surface is the initial step in transducing an extracellular signal into an intracellular response. This proximal event in WNT signal transduction with (cell-specific) ligand-receptor interactions is of great interest as a potential target for pharmacological intervention. In this review we highlight the diverse expression of various WNT receptors and co-receptors in the aforementioned chronic lung diseases and discuss the currently available biologicals and pharmacological tools to modify proximal WNT signalling.
Collapse
Affiliation(s)
- Wioletta Skronska-Wasek
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Hoeke Abele Baarsma
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
42
|
Wujak L, Schnieder J, Schaefer L, Wygrecka M. LRP1: A chameleon receptor of lung inflammation and repair. Matrix Biol 2017; 68-69:366-381. [PMID: 29262309 DOI: 10.1016/j.matbio.2017.12.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
The lung displays a remarkable capability to regenerate following injury. Considerable effort has been made thus far to understand the cardinal processes underpinning inflammation and reconstruction of lung tissue. However, the factors determining the resolution or persistence of inflammation and efficient wound healing or aberrant remodeling remain largely unknown. Low density lipoprotein receptor-related protein 1 (LRP1) is an endocytic/signaling cell surface receptor which controls cellular and molecular mechanisms driving the physiological and pathological inflammatory reactions and tissue remodeling in several organs. In this review, we will discuss the impact of LRP1 on the consecutive steps of the inflammatory response and its role in the balanced tissue repair and aberrant remodeling in the lung.
Collapse
Affiliation(s)
- Lukasz Wujak
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Jennifer Schnieder
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Liliana Schaefer
- Goethe University School of Medicine, University Hospital, Theodor-Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany; Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
43
|
Chukkapalli SS, Easwaran M, Rivera-Kweh MF, Velsko IM, Ambadapadi S, Dai J, Larjava H, Lucas AR, Kesavalu L. Sequential colonization of periodontal pathogens in induction of periodontal disease and atherosclerosis in LDLRnull mice. Pathog Dis 2017; 75:ftx003. [PMID: 28104616 DOI: 10.1093/femspd/ftx003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/13/2017] [Indexed: 12/13/2022] Open
Abstract
Periodontal disease (PD) and atherosclerotic vascular disease (ASVD) are both chronic inflammatory diseases with a polymicrobial etiology and have been epidemiologically associated. The purpose is to examine whether periodontal bacteria that infect the periodontium can also infect vascular tissues and enhance pre-existing early aortic atherosclerotic lesions in LDLRnull mice. Mice were orally infected with intermediate bacterial colonizer Fusobacterium nucleatum for the first 12 weeks followed by late bacterial colonizers (Porphyromonas gingivalis, Treponema denticola and Tannerella forsythia) for the remaining 12 weeks mimicking the human oral microbiota ecological colonization. Genomic DNA from all four bacterial was detected in gingival plaque by PCR, consistently demonstrating infection of mouse gingival surfaces. Infected mice had significant levels of IgG and IgM antibodies, alveolar bone resorption, and showed apical migration of junctional epithelium revealing the induction of PD. These results support the ability of oral bacteria to cause PD in mice. Detection of bacterial genomic DNA in systemic organs indicates hematogenous dissemination from the gingival pockets. Bacterial infection did not alter serum lipid fractions or serum amyloid A levels and did not induce aortic atherosclerotic plaque. This is the first study examining the causal role of periodontal bacteria in induction of ASVD in LDLRnull mice.
Collapse
Affiliation(s)
- Sasanka S Chukkapalli
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA
| | - Meena Easwaran
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA
| | - Mercedes F Rivera-Kweh
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA
| | - Irina M Velsko
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA
| | - Sriram Ambadapadi
- Biodesign Institute, Arizona state University, Tempe, AZ 85287-5001, USA
| | - Jiayin Dai
- Division of Periodontics and Dental Hygiene, University of British Columbia, Vancouver, BCV6T 1Z4, Canada
| | - Hannu Larjava
- Division of Periodontics and Dental Hygiene, University of British Columbia, Vancouver, BCV6T 1Z4, Canada
| | - Alexandra R Lucas
- Biodesign Institute, Arizona state University, Tempe, AZ 85287-5001, USA
| | - Lakshmyya Kesavalu
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA.,Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610-0424, USA
| |
Collapse
|
44
|
Tham DKL, Moukhles H. Determining Cell-surface Expression and Endocytic Rate of Proteins in Primary Astrocyte Cultures Using Biotinylation. J Vis Exp 2017. [PMID: 28715371 DOI: 10.3791/55974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Cell-surface proteins mediate a wide array of functions. In many cases, their activity is regulated by endocytic processes that modulate their levels at the plasma membrane. Here, we present detailed protocols for 2 methods that facilitate the study of such processes, both of which are based on the principle of the biotinylation of cell-surface proteins. The first is designed to allow for the semi-quantitative determination of the relative levels of a particular protein at the cell-surface. In it, the lysine residues of the plasma membrane proteins of cells are first labeled with a biotin moiety. Once the cells are lysed, these proteins may then be specifically precipitated via the use of agarose-immobilized streptavidin by exploiting the natural affinity of the latter for biotin. The proteins isolated in such a manner may then be analyzed via a standard western blotting approach. The second method provides a means of determining the endocytic rate of a particular cell-surface target over a period of time. Cell-surface proteins are first modified with a biotin derivative containing a cleavable disulfide bond. The cells are then shifted back to normal culture conditions, which causes the endocytic uptake of a proportion of biotinylated proteins. Next, the disulfide bonds of non-internalized biotin groups are reduced using the membrane-impermeable reducing agent glutathione. Via this approach, endocytosed proteins may thus be isolated and quantified with a high degree of specificity.
Collapse
Affiliation(s)
- Daniel Kai Long Tham
- Department of Cellular and Physiological Sciences, University of British Columbia
| | - Hakima Moukhles
- Department of Cellular and Physiological Sciences, University of British Columbia;
| |
Collapse
|
45
|
Elsafadi M, Manikandan M, Alajez NM, Hamam R, Dawud RA, Aldahmash A, Iqbal Z, Alfayez M, Kassem M, Mahmood A. MicroRNA-4739 regulates osteogenic and adipocytic differentiation of immortalized human bone marrow stromal cells via targeting LRP3. Stem Cell Res 2017; 20:94-104. [PMID: 28340487 DOI: 10.1016/j.scr.2017.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/25/2017] [Accepted: 03/01/2017] [Indexed: 12/16/2022] Open
Abstract
Understanding the regulatory networks underlying lineage differentiation and fate determination of human bone marrow stromal cells (hBMSC) is a prerequisite for their therapeutic use. The goal of the current study was to unravel the novel role of the low-density lipoprotein receptor-related protein 3 (LRP3) in regulating the osteogenic and adipogenic differentiation of immortalized hBMSCs. Gene expression profiling revealed significantly higher LRP3 levels in the highly osteogenic hBMSC clone imCL1 than in the less osteogenic clone imCL2, as well as a significant upregulation of LRP3 during the osteogenic induction of the imCL1 clone. Data from functional and gene expression assays demonstrated the role of LRP3 as a molecular switch promoting hBMSC lineage differentiation into osteoblasts and inhibiting differentiation into adipocytes. Interestingly, microRNA (miRNA) expression profiling identified miR-4739 as the most under-represented miRNA (-36.11 fold) in imCL1 compared to imCL2. The TargetScan prediction algorithm, combined with functional and biochemical assays, identified LRP3 mRNA as a novel target of miR-4739, with a single potential binding site for miR-4739 located in the LRP3 3' UTR. Regulation of LRP3 expression by miR-4739 was subsequently confirmed by qRT-PCR, western blotting, and luciferase assays. Over-expression of miR-4739 mimicked the effects of LRP3 knockdown on promoting adipogenic and suppressing osteogenic differentiation of hBMSCs. Hence, we report for the first time a novel biological role for the LRP3/hsa-miR-4739 axis in balancing osteogenic and adipocytic differentiation of hBMSCs. Our data support the potential utilization of miRNA-based therapies in regenerative medicine.
Collapse
Affiliation(s)
- Mona Elsafadi
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia; KMEB, Department of Endocrinology, University Hospital of Odense, University of Southern Denmark, Winslowsparken 25.1, DK-5000 Odense C, Denmark.
| | - Muthurangan Manikandan
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia
| | - Nehad M Alajez
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia.
| | - Rimi Hamam
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia
| | - Raed Abu Dawud
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia
| | - Abdullah Aldahmash
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia; Prince Naif Health Research Center, King Saud University, Riyadh 11461, Saudi Arabia.
| | - Zafar Iqbal
- Department of Basic Sciences, College of applied medical sciences, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), National Guard Health Affairs, Al Ahsa, Saudi Arabia
| | - Musaad Alfayez
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia.
| | - Moustapha Kassem
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia; KMEB, Department of Endocrinology, University Hospital of Odense, University of Southern Denmark, Winslowsparken 25.1, DK-5000 Odense C, Denmark.
| | - Amer Mahmood
- Stem Cell Unit, Department of Anatomy, College of Medicine,King Saud University, Riyadh 11461, Saudi Arabia.
| |
Collapse
|
46
|
Pohlkamp T, Wasser CR, Herz J. Functional Roles of the Interaction of APP and Lipoprotein Receptors. Front Mol Neurosci 2017; 10:54. [PMID: 28298885 PMCID: PMC5331069 DOI: 10.3389/fnmol.2017.00054] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/16/2017] [Indexed: 11/24/2022] Open
Abstract
The biological fates of the key initiator of Alzheimer’s disease (AD), the amyloid precursor protein (APP), and a family of lipoprotein receptors, the low-density lipoprotein (LDL) receptor-related proteins (LRPs) and their molecular roles in the neurodegenerative disease process are inseparably interwoven. Not only does APP bind tightly to the extracellular domains (ECDs) of several members of the LRP group, their intracellular portions are also connected through scaffolds like the one established by FE65 proteins and through interactions with adaptor proteins such as X11/Mint and Dab1. Moreover, the ECDs of APP and LRPs share common ligands, most notably Reelin, a regulator of neuronal migration during embryonic development and modulator of synaptic transmission in the adult brain, and Agrin, another signaling protein which is essential for the formation and maintenance of the neuromuscular junction (NMJ) and which likely also has critical, though at this time less well defined, roles for the regulation of central synapses. Furthermore, the major independent risk factors for AD, Apolipoprotein (Apo) E and ApoJ/Clusterin, are lipoprotein ligands for LRPs. Receptors and ligands mutually influence their intracellular trafficking and thereby the functions and abilities of neurons and the blood-brain-barrier to turn over and remove the pathological product of APP, the amyloid-β peptide. This article will review and summarize the molecular mechanisms that are shared by APP and LRPs and discuss their relative contributions to AD.
Collapse
Affiliation(s)
- Theresa Pohlkamp
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Catherine R Wasser
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA; Department of Neuroscience, UT Southwestern Medical CenterDallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
47
|
El Gaamouch F, Jing P, Xia J, Cai D. Alzheimer's Disease Risk Genes and Lipid Regulators. J Alzheimers Dis 2017; 53:15-29. [PMID: 27128373 DOI: 10.3233/jad-160169] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Brain lipid homeostasis plays an important role in Alzheimer's disease (AD) and other neurodegenerative disorders. Aggregation of amyloid-β peptide is one of the major events in AD. The complex interplay between lipids and amyloid-β accumulation has been intensively investigated. The proportions of lipid components including phospholipids, sphingolipids, and cholesterol are roughly similar across different brain regions under physiological conditions. However, disruption of brain lipid homeostasis has been described in AD and implicated in disease pathogenesis. Moreover, studies suggest that analysis of lipid composition in plasma and cerebrospinal fluid could improve our understanding of the disease development and progression, which could potentially serve as disease biomarkers and prognostic indicators for AD therapies. Here, we summarize the functional roles of AD risk genes and lipid regulators that modulate brain lipid homeostasis including different lipid species, lipid complexes, and lipid transporters, particularly their effects on amyloid processing, clearance, and aggregation, as well as neuro-toxicities that contribute to AD pathogenesis.
Collapse
Affiliation(s)
- Farida El Gaamouch
- James J Peters VA Medical Center, Research & Development, Bronx, NY, USA.,Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ping Jing
- The Central Hospital of Wuhan, China
| | | | - Dongming Cai
- James J Peters VA Medical Center, Research & Development, Bronx, NY, USA.,Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Central Hospital of Wuhan, China
| |
Collapse
|
48
|
Bochkov YA, Gern JE. Rhinoviruses and Their Receptors: Implications for Allergic Disease. Curr Allergy Asthma Rep 2016; 16:30. [PMID: 26960297 DOI: 10.1007/s11882-016-0608-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Human rhinoviruses (RVs) are picornaviruses that can cause a variety of illnesses including the common cold, lower respiratory tract illnesses such as bronchitis and pneumonia, and exacerbations of asthma. RVs are classified into three species, RV-A, B, and C, which include over 160 types. They utilize three major types of cellular membrane glycoproteins to gain entry into the host cell: intercellular adhesion molecule 1 (ICAM-1) (the majority of RV-A and all RV-B), low-density lipoprotein receptor (LDLR) family members (12 RV-A types), and cadherin-related family member 3 (CDHR3) (RV-C). CDHR3 is a member of cadherin superfamily of transmembrane proteins with yet unknown biological function, and there is relatively little information available about the mechanisms of RV-C interaction with CDHR3. A coding single nucleotide polymorphism (rs6967330) in CDHR3 could promote RV-C infections and illnesses in infancy, which could in turn adversely affect the developing lung to increase the risk of asthma. Further studies are needed to determine how RV infections contribute to pathogenesis of asthma and to develop the optimal treatment approach to control asthma exacerbations.
Collapse
Affiliation(s)
- Yury A Bochkov
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI, 53792, USA.
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI, 53792, USA.,Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave, Madison, WI, 53792, USA
| |
Collapse
|
49
|
Mesd extrinsically promotes phagocytosis by retinal pigment epithelial cells. Cell Biol Toxicol 2016; 32:347-58. [PMID: 27184668 DOI: 10.1007/s10565-016-9339-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/09/2016] [Indexed: 01/09/2023]
Abstract
Phagocytosis is a critical process to maintain tissue homeostasis. In the retina, photoreceptor cells renew their photoexcitability by shedding photoreceptor outer segments (POSs) in a diurnal rhythm. Shed POSs are phagocytosed by retinal pigment epithelial (RPE) cells to prevent debris accumulation, retinal degeneration, and blindness. Phagocytosis ligands are the key to understanding how RPE recognizes shed POSs. Here, we characterized mesoderm development candidate 2 (Mesd or Mesdc2), an endoplasmic reticulum (ER) chaperon for low-density lipoprotein receptor-related proteins (LRPs), to extrinsically promote RPE phagocytosis. The results showed that Mesd stimulated phagocytosis of fluorescence-labeled POS vesicles by D407 RPE cells. Ingested POSs were partially degraded within 3 h in some RPE cells to dispense undegradable fluorophore throughout the cytoplasm. Internalized POSs were colocalized with phagosome biomarker Rab7, suggesting that Mesd-mediated engulfment is involved in a phagocytosis pathway. Mesd also facilitated phagocytosis of POSs by primary RPE cells. Mesd bound to unknown phagocytic receptor(s) on RPE cells. Mesd was detected in the cytoplasm, but not nuclei, of different retinal layers and is predominantly expressed in the ER-free cellular compartment of POSs. Mesd was not secreted into medium from healthy cells but passively released from apoptotic cells with increased membrane permeability. Released Mesd selectively bound to the surface of POS vesicles and apoptotic cells, but not healthy cells. These results suggest that Mesd may be released from and bind to shed POSs to facilitate their phagocytic clearance.
Collapse
|
50
|
Cenarro A, Etxebarria A, de Castro-Orós I, Stef M, Bea AM, Palacios L, Mateo-Gallego R, Benito-Vicente A, Ostolaza H, Tejedor T, Martín C, Civeira F. The p.Leu167del Mutation in APOE Gene Causes Autosomal Dominant Hypercholesterolemia by Down-regulation of LDL Receptor Expression in Hepatocytes. J Clin Endocrinol Metab 2016; 101:2113-21. [PMID: 27014949 DOI: 10.1210/jc.2015-3874] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
CONTEXT The p.Leu167del mutation in the APOE gene has been associated with hyperlipidemia. OBJECTIVES Our objective was to determine the frequency of p.Leu167del mutation in APOE gene in subjects with autosomal dominant hypercholesterolemia (ADH) in whom LDLR, APOB, and PCSK9 mutations had been excluded and to identify the mechanisms by which this mutant apo E causes hypercholesterolemia. DESIGN The APOE gene was analyzed in a case-control study. SETTING The study was conducted at a University Hospital Lipid Clinic. PATIENTS OR OTHER PARTICIPANTS Two groups (ADH, 288 patients; control, 220 normolipidemic subjects) were included. INTERVENTION We performed sequencing of APOE gene and proteomic and cellular experiments. MAIN OUTCOME MEASURE To determine the frequency of the p.Leu167del mutation and the mechanism by which it causes hypercholesterolemia. RESULTS In the ADH group, nine subjects (3.1%) were carriers of the APOE c.500_502delTCC, p.Leu167del mutation, cosegregating with hypercholesterolemia in studied families. Proteomic quantification of wild-type and mutant apo E in very low-density lipoprotein (VLDL) from carrier subjects revealed that apo E3 is almost a 5-fold increase compared to mutant apo E. Cultured cell studies revealed that VLDL from mutation carriers had a significantly higher uptake by HepG2 and THP-1 cells compared to VLDL from subjects with E3/E3 or E2/E2 genotypes. Transcriptional down-regulation of LDLR was also confirmed. CONCLUSIONS p.Leu167del mutation in APOE gene is the cause of hypercholesterolemia in the 3.1% of our ADH subjects without LDLR, APOB, and PCSK9 mutations. The mechanism by which this mutation is associated to ADH is that VLDL carrying the mutant apo E produces LDLR down-regulation, thereby raising plasma low-density lipoprotein cholesterol levels.
Collapse
Affiliation(s)
- Ana Cenarro
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Aitor Etxebarria
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Isabel de Castro-Orós
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Marianne Stef
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Ana M Bea
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Lourdes Palacios
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Rocío Mateo-Gallego
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Asier Benito-Vicente
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Helena Ostolaza
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Teresa Tejedor
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - César Martín
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis (A.C., I.d.C.-O., A.M.B., R.M.-G., F.C.), Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain; Unidad de Biofísica (Consejo Superior de Investigaciones Científicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea) and Departamento de Bioquímica y Biología Molecular (A.E., A.B.-V., H.O., C.M.), Universidad del País Vasco/Euskal Herriko Unibertsitatea, Bilbao, Spain; Progenika Biopharma (M.S., L.P.), a Grifols Company, Derio, Spain; and Departamento de Anatomía (T.T.), Embriología y Genética, Universidad de Zaragoza, Spain
| |
Collapse
|