1
|
Cloutier G, Beaulieu JF. Reconsideration of the laminin receptor 67LR in colorectal cancer cells. BIOMOLECULES & BIOMEDICINE 2024; 24:1117-1132. [PMID: 38606907 PMCID: PMC11378999 DOI: 10.17305/bb.2024.10323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
The 67 kDa laminin receptor (67LR) was identified as the first laminin receptor shown to be involved in the carcinogenesis of various cancers, including colorectal cancer. While the exact composition of this 67 kDa receptor remains unknown, it has been reported to be formed by the 37 kDa ribosomal protein SA (RPSA) covalently attached to another unidentified protein. The goal of this study was to clarify the molecular structure of 67LR to enhance our understanding of its role in malignancies. Using cell fractionation of colorectal cancer cells, the 67 kDa immunoreactive protein corresponding to 67LR was found in the soluble protein fraction, while some of the 37 kDa RPSA exhibited plasma membrane-like properties. Proteomic analysis of the 67 kDa fraction revealed the absence of RPSA but identified the β-galactosidase-related 67 kDa elastin-binding protein (67EBP), another laminin binding receptor which presents amino acid sequence similarities that can explain the immune cross reactivity with RPSA. The downregulation of β-galactosidase through short hairpin RNA (shRNA) led to a reduction in both 67LR and 67EBP immunoreactive proteins, confirming the misidentification of 67LR and 67EBP in colorectal cancer cells. Based on these findings, we propose to redefine the 67LR as the RPSA-containing laminin receptor (RCLR) to avoid confusion with the 67EBP.
Collapse
Affiliation(s)
- Gabriel Cloutier
- Department of Immunology and Cell Biology, Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Jean-François Beaulieu
- Department of Immunology and Cell Biology, Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
2
|
Haddad R, Sadeh O, Ziv T, Erlich I, Haimovich-Caspi L, Shemesh A, van der Velden J, Kehat I. Localized translation and sarcomere maintenance requires ribosomal protein SA in mice. J Clin Invest 2024; 134:e174527. [PMID: 38743494 PMCID: PMC11213506 DOI: 10.1172/jci174527] [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: 08/11/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Cardiomyocyte sarcomeres contain localized ribosomes, but the factors responsible for their localization and the significance of localized translation are unknown. Using proximity labeling, we identified ribosomal protein SA (RPSA) as a Z-line protein. In cultured cardiomyocytes, the loss of RPSA led to impaired local protein translation and reduced sarcomere integrity. By employing CAS9-expressing mice, along with adeno-associated viruses expressing CRE recombinase and single-guide RNAs targeting Rpsa, we knocked out Rpsa in vivo and observed mislocalization of ribosomes and diminished local translation. These genetic mosaic mice with Rpsa knockout in a subset of cardiomyocytes developed dilated cardiomyopathy, featuring atrophy of RPSA-deficient cardiomyocytes, compensatory hypertrophy of unaffected cardiomyocytes, left ventricular dilation, and impaired contractile function. We demonstrated that RPSA C-terminal domain is sufficient for localization to the Z-lines and that if the microtubule network is disrupted RPSA loses its sarcomeric localization. These findings highlight RPSA as a ribosomal factor essential for ribosome localization to the Z-line, facilitating local translation and sarcomere maintenance.
Collapse
Affiliation(s)
- Rami Haddad
- Rappaport Family Institute for Research in the Medical Sciences and The Ruth and Bruce Rappaport Faculty of Medicine
| | - Omer Sadeh
- Rappaport Family Institute for Research in the Medical Sciences and The Ruth and Bruce Rappaport Faculty of Medicine
| | - Tamar Ziv
- Smoler Proteomics Center, Lorry I. Lokey Interdisciplinary Center for Life Sciences and Engineering, and
| | - Itai Erlich
- Rappaport Family Institute for Research in the Medical Sciences and The Ruth and Bruce Rappaport Faculty of Medicine
| | - Lilac Haimovich-Caspi
- Rappaport Family Institute for Research in the Medical Sciences and The Ruth and Bruce Rappaport Faculty of Medicine
| | - Ariel Shemesh
- Biomedical Core Facility at the Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Izhak Kehat
- Rappaport Family Institute for Research in the Medical Sciences and The Ruth and Bruce Rappaport Faculty of Medicine
| |
Collapse
|
3
|
Cuttler K, Bignoux MJ, Otgaar TC, Chigumba S, Ferreira E, Weiss SFT. LRP::FLAG Reduces Phosphorylated Tau Levels in Alzheimer's Disease Cell Culture Models. J Alzheimers Dis 2021; 76:753-768. [PMID: 32568204 DOI: 10.3233/jad-200244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque and neurofibrillary tangle formation, respectively. Neurofibrillary tangles form as a result of the intracellular accumulation of hyperphosphorylated tau. Telomerase activity and levels of the human reverse transcriptase (hTERT) subunit of telomerase are significantly decreased in AD. Recently, it has been demonstrated that the 37 kDa/67 kDa laminin receptor (LRP/LR) interacts with telomerase and is implicated in Aβ pathology. Since both LRP/LR and telomerase are known to play a role in the Aβ facet of AD, we hypothesized that they might also play a role in tauopathy. OBJECTIVE This study aimed to determine if LRP/LR has a relationship with tau and whether overexpression of LRP::FLAG has an effect on tauopathy-related proteins. METHODS We employed confocal microscopy and FRET to determine whether LRP/LR and tau co-localize and interact. LRP::FLAG overexpression in HEK-293 and SH-SY5Y cells as well as analysis of tauopathy-related proteins was assessed by western blotting. RESULTS We demonstrate that LRP/LR co-localizes with tau in the perinuclear cell compartment and confirmed a direct interaction between LRP/LR and tau in HEK-293 cells. Overexpression of LRP::FLAG in HEK-293 and SH-SY5Y cells decreased total and phosphorylated tau levels with a concomitant decrease in PrPc levels, a tauopathy-related protein. LRP::FLAG overexpression also resulted in increased hTERT levels. CONCLUSION This data suggest that LRP/LR extends its role in AD through a direct interaction with tau, and recommend LRP::FLAG as a possible alternative AD therapeutic via decreasing phosphorylated tau levels.
Collapse
Affiliation(s)
- Katelyn Cuttler
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa.,Present Address: Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Republic of South Africa
| | - Monique J Bignoux
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Tyrone C Otgaar
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Stephanie Chigumba
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Eloise Ferreira
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Stefan F T Weiss
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| |
Collapse
|
4
|
Shan F, Mei S, Zhang J, Zhang X, Xu C, Liao S, Tu X. A telomerase subunit homolog La protein from
Trypanosoma brucei
plays an essential role in ribosomal biogenesis. FEBS J 2019; 286:3129-3147. [DOI: 10.1111/febs.14853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/23/2019] [Accepted: 04/13/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Fangzhen Shan
- Hefei National Laboratory for Physical Science at Microscale School of Life Science University of Science and Technology of China Hefei China
| | - Song Mei
- Key Laboratory of Tropical Forest Ecology Xishuangbanna Tropical Botanical Garden Chinese Academy of Sciences Kunming China
| | - Jiahai Zhang
- Hefei National Laboratory for Physical Science at Microscale School of Life Science University of Science and Technology of China Hefei China
| | | | - Chao Xu
- Hefei National Laboratory for Physical Science at Microscale School of Life Science University of Science and Technology of China Hefei China
| | - Shanhui Liao
- Hefei National Laboratory for Physical Science at Microscale School of Life Science University of Science and Technology of China Hefei China
| | - Xiaoming Tu
- Hefei National Laboratory for Physical Science at Microscale School of Life Science University of Science and Technology of China Hefei China
| |
Collapse
|
5
|
Griffin JN, Sondalle SB, Robson A, Mis EK, Griffin G, Kulkarni SS, Deniz E, Baserga SJ, Khokha MK. RPSA, a candidate gene for isolated congenital asplenia, is required for pre-rRNA processing and spleen formation in Xenopus. Development 2018; 145:145/20/dev166181. [PMID: 30337486 DOI: 10.1242/dev.166181] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 09/13/2018] [Indexed: 12/14/2022]
Abstract
A growing number of tissue-specific inherited disorders are associated with impaired ribosome production, despite the universal requirement for ribosome function. Recently, mutations in RPSA, a protein component of the small ribosomal subunit, were discovered to underlie approximately half of all isolated congenital asplenia cases. However, the mechanisms by which mutations in this ribosome biogenesis factor lead specifically to spleen agenesis remain unknown, in part due to the lack of a suitable animal model for study. Here we reveal that RPSA is required for normal spleen development in the frog, Xenopus tropicalis Depletion of Rpsa in early embryonic development disrupts pre-rRNA processing and ribosome biogenesis, and impairs expression of the key spleen patterning genes nkx2-5, bapx1 and pod1 in the spleen anlage. Importantly, we also show that whereas injection of human RPSA mRNA can rescue both pre-rRNA processing and spleen patterning, injection of human mRNA bearing a common disease-associated mutation cannot. Together, we present the first animal model of RPSA-mediated asplenia and reveal a crucial requirement for RPSA in pre-rRNA processing and molecular patterning during early Xenopus development.
Collapse
Affiliation(s)
- John N Griffin
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Samuel B Sondalle
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Andrew Robson
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Emily K Mis
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Gerald Griffin
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Saurabh S Kulkarni
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Engin Deniz
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Susan J Baserga
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA .,Departments of Molecular Biophysics and Biochemistry, and Therapeutic Radiology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA .,Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06510, USA
| |
Collapse
|
6
|
Butò S, Ghirelli C, Aiello P, Tagliabue E, Ardini E, Magnifico A, Montuori N, Sobel ME, Colnaghi MI, Ménard S. Production and Characterization of Monoclonal Antibodies Directed against the Laminin Receptor Precursor. Int J Biol Markers 2018; 12:1-5. [PMID: 9176710 DOI: 10.1177/172460089701200101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The 67-kDa laminin receptor (67LR) is an important tumor marker whose molecular structure has not yet been fully elucidated. To shed new light on this molecule, we raised a series of eight new monoclonal antibodies, designated MPLR1 to 8, directed against the 37-kDa recombinant laminin receptor precursor (37LRP). Cross-competition experiments demonstrated that the epitopes recognized by MPLR2, 4 and 5 partially overlap, since MPLR4 and 5 compete with labelled MPLR2 for the binding to recombinant 37LRP. These three antibodies belong to the IgG1 class, whereas the other ones are all IgM. Presumably due to the fact that they are directed against partially unfolded antigenic determinants expressed on the recombinant protein, MPLRs did not recognize the native protein. Indeed, they showed no reactivity at the membrane level in cytofluorimetric analysis and they did not work in immunoprecipitation experiments. In contrast, these reagents are valuable tools in immunoblotting, since they clearly identify a 67-kDa protein (the mature laminin receptor) in addition to the 37-kDa precursor form. MPLRs are thus a new powerful tool which could help in the characterization of the still enigmatic 67LR molecule.
Collapse
Affiliation(s)
- S Butò
- Division of Experimental Oncology E, Istituto Nazionale Tumori, Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Chetty CJ, Ferreira E, Jovanovic K, Weiss SFT. Knockdown of LRP/LR induces apoptosis in pancreatic cancer and neuroblastoma cells through activation of caspases. Exp Cell Res 2017; 360:264-272. [PMID: 28899658 DOI: 10.1016/j.yexcr.2017.09.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
The 37kDa/67kDa laminin receptor (LRP/LR) serves various physiological and pathological roles such as enhancing tumour-related processes including metastasis, angiogenesis, cellular viability and telomerase activation in cancerous cell lines. The present study investigates the effect of siRNA mediated downregulation of LRP/LR on pancreatic cancer (AsPC-1) and neuroblastoma (IMR-32) cells. MTT and BrdU assays revealed that siRNA mediated downregulation of LRP resulted in a significant reduction in cell viability and cell proliferation. In addition, knock-down of LRP resulted in phosphatidylserine externalization, diminished nuclear integrity and significantly enhanced caspase-3 activity, which is indicative of apoptosis. LRP downregulation resulted in a significant increase in caspase-8 activity in IMR-32 cells and enhanced caspase-8 and 9 activity in AsPC-1 cells. These data recommend siRNA mediated knock-down of LRP as a potential therapeutic avenue for the treatment of pancreatic cancer and neuroblastoma.
Collapse
Affiliation(s)
- Carryn J Chetty
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, Republic of South Africa
| | - Eloise Ferreira
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, Republic of South Africa
| | - Katarina Jovanovic
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, Republic of South Africa
| | - Stefan F T Weiss
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, Republic of South Africa.
| |
Collapse
|
8
|
Aryantini NPD, Kondoh D, Nishiyama K, Yamamoto Y, Mukai T, Sujaya IN, Urashima T, Fukuda K. Anchorless cell surface proteins function as laminin-binding adhesins in Lactobacillus rhamnosus FSMM22. FEMS Microbiol Lett 2017; 364:3064890. [PMID: 28333282 DOI: 10.1093/femsle/fnx056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/08/2017] [Indexed: 11/12/2022] Open
Abstract
Anchorless cell surface proteins (CSPs) were extracted with 1 M lithium chloride solution from Lactobacillus rhamnosus FSMM22. Loss of the anchorless CSPs resulted in a 2-fold decrease in FSMM22 cells bound to a constitutive extracellular matrix glycoprotein, laminin, in vitro. DNA-binding protein HU, glyceraldehyde-3-phosphate dehydrogenase, lactate dehydrogenase and 30S ribosomal protein S19 (RpsS) were identified by mass spectrometry in the extract as laminin-binding adhesins. Among the four proteins, RpsS was immunohistochemically confirmed to exist on the cell surface. Our findings strongly suggest that anchorless CSPs can enhance bacterial adhesion to the host.
Collapse
Affiliation(s)
- Ni Putu Desy Aryantini
- Department of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Daisuke Kondoh
- Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido 080-8555, Japan
| | - Keita Nishiyama
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan
| | - Yuji Yamamoto
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan
| | - Takao Mukai
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan
| | - I Nengah Sujaya
- Integrated Laboratory for Bioscience and Biotechnology, Udayana University, Bukit Jimbaran Campus, Badung, Bali 80361, Indonesia
| | - Tadasu Urashima
- Department of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Kenji Fukuda
- Department of Animal and Food Hygiene, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
9
|
Discovery of new small molecules inhibiting 67 kDa laminin receptor interaction with laminin and cancer cell invasion. Oncotarget 2016; 6:18116-33. [PMID: 26062445 PMCID: PMC4627239 DOI: 10.18632/oncotarget.4016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/18/2015] [Indexed: 01/23/2023] Open
Abstract
The 67 kDa laminin receptor (67LR) is a non-integrin receptor for laminin (LM) that derives from a 37 kDa precursor (37LRP). 67LR expression is increased in neoplastic cells and correlates with an enhanced invasive and metastatic potential. We used structure-based virtual screening (SB-VS) to search for 67LR inhibitory small molecules, by focusing on a 37LRP sequence, the peptide G, able to specifically bind LM. Forty-six compounds were identified and tested on HEK-293 cells transfected with 37LRP/67LR (LR-293 cells). One compound, NSC47924, selectively inhibited LR-293 cell adhesion to LM with IC50 and Ki values of 19.35 and 2.45 μmol/L. NSC47924 engaged residues W176 and L173 of peptide G, critical for specific LM binding. Indeed, NSC47924 inhibited in vitro binding of recombinant 37LRP to both LM and its YIGSR fragment. NSC47924 also impaired LR-293 cell migration to LM and cell invasion. A subsequent hierarchical similarity search with NSC47924 led to the identification of additional four compounds inhibiting LR-293 cell binding to LM: NSC47923, NSC48478, NSC48861, and NSC48869, with IC50 values of 1.99, 1.76, 3.4, and 4.0 μmol/L, respectively, and able to block in vitro cancer cell invasion. These compounds are promising scaffolds for future drug design and discovery efforts in cancer progression.
Collapse
|
10
|
Mazur AJ, Radaszkiewicz T, Makowiecka A, Malicka-Błaszkiewicz M, Mannherz HG, Nowak D. Gelsolin interacts with LamR, hnRNP U, nestin, Arp3 and β-tubulin in human melanoma cells as revealed by immunoprecipitation and mass spectrometry. Eur J Cell Biol 2016; 95:26-41. [DOI: 10.1016/j.ejcb.2015.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/08/2015] [Accepted: 11/04/2015] [Indexed: 01/25/2023] Open
|
11
|
Graifer D, Karpova G. Roles of ribosomal proteins in the functioning of translational machinery of eukaryotes. Biochimie 2015; 109:1-17. [DOI: 10.1016/j.biochi.2014.11.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 11/18/2014] [Indexed: 11/16/2022]
|
12
|
DiGiacomo V, Meruelo D. Looking into laminin receptor: critical discussion regarding the non-integrin 37/67-kDa laminin receptor/RPSA protein. Biol Rev Camb Philos Soc 2015; 91:288-310. [PMID: 25630983 DOI: 10.1111/brv.12170] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/04/2014] [Accepted: 12/08/2014] [Indexed: 02/06/2023]
Abstract
The 37/67-kDa laminin receptor (LAMR/RPSA) was originally identified as a 67-kDa binding protein for laminin, an extracellular matrix glycoprotein that provides cellular adhesion to the basement membrane. LAMR has evolutionary origins, however, as a 37-kDa RPS2 family ribosomal component. Expressed in all domains of life, RPS2 proteins have been shown to have remarkably diverse physiological roles that vary across species. Contributing to laminin binding, ribosome biogenesis, cytoskeletal organization, and nuclear functions, this protein governs critical cellular processes including growth, survival, migration, protein synthesis, development, and differentiation. Unsurprisingly given its purview, LAMR has been associated with metastatic cancer, neurodegenerative disease and developmental abnormalities. Functioning in a receptor capacity, this protein also confers susceptibility to bacterial and viral infection. LAMR is clearly a molecule of consequence in human disease, directly mediating pathological events that make it a prime target for therapeutic interventions. Despite decades of research, there are still a large number of open questions regarding the cellular biology of LAMR, the nature of its ability to bind laminin, the function of its intrinsically disordered C-terminal region and its conversion from 37 to 67 kDa. This review attempts to convey an in-depth description of the complexity surrounding this multifaceted protein across functional, structural and pathological aspects.
Collapse
Affiliation(s)
- Vincent DiGiacomo
- Department of Pathology, New York University School of Medicine, 180 Varick Street, New York, NY 10014, U.S.A
| | - Daniel Meruelo
- Department of Pathology, New York University School of Medicine, 180 Varick Street, New York, NY 10014, U.S.A.,NYU Cancer Institute, 550 First Avenue, New York, NY 10016, U.S.A.,NYU Gene Therapy Center, 550 First Avenue, New York, NY 10016, U.S.A
| |
Collapse
|
13
|
Bolze A. [Connecting isolated congenital asplenia to the ribosome]. Biol Aujourdhui 2014; 208:289-98. [PMID: 25840456 DOI: 10.1051/jbio/2015001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Indexed: 11/14/2022]
Abstract
Isolated congenital asplenia is characterized by the absence of a spleen at birth without any other developmental defect. Isolated congenital asplenia is a rare and life-threatening disease that predisposes patients to severe bacterial infections. The first and main genetic etiology of isolated congenital asplenia was discovered in 2013. Mutations in the gene RPSA, which encodes ribosomal protein SA, cause more than half of the cases of isolated congenital asplenia. These disease-causing mutations lead to haploinsufficiency of RPSA. Haploinsufficiency of genes encoding other ribosomal proteins have been reported to cause other developmental defects in humans, and in model organisms like the fly or the mouse. About half of the patients with Diamond-Blackfan anemia, which is a well-characterized ribosomopathy, present developmental defects such as craniofacial defects, cardiac defects or thumb abnormalities. The mechanism of pathogenesis linking mutations in ribosomal proteins, which are highly and ubiquitously expressed, to specific developmental defects remains to be elucidated. One hypothesis is that the ribosome, and ribosomal proteins in particular, regulate the expression of specific genes during development.
Collapse
|
14
|
Parkin induces upregulation of 40S ribosomal protein SA and posttranslational modification of cytokeratins 8 and 18 in human cervical cancer cells. Appl Biochem Biotechnol 2013; 171:1630-8. [PMID: 23990477 DOI: 10.1007/s12010-013-0443-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/06/2013] [Indexed: 10/26/2022]
Abstract
Parkin was originally identified as a protein associated with Parkinson's disease. Recently, numerous research studies have suggested that parkin acts as a tumor suppressor. In accordance with these studies, we previously reported that overexpression of parkin in HeLa cells induced growth inhibition. To elucidate possible mechanisms by which parkin may inhibit cell growth, HeLa cells were infected with adenoviruses expressing either the parkin gene or adenovirus alone for 72 h and a total proteomic analysis was performed using 2-D gel electrophoresis followed by LC-MS/MS. We identified three proteins whose expression changed between the two groups: the 40S ribosomal protein SA (RPSA) was downregulated in parkin virus-infected cells, and cytokeratins 8 and 18 exhibited an acid shift in pI value without a change in molecular weight, suggesting that these proteins became phosphorylated in parkin virus-infected cells. The changes in these three proteins were first observed at 60 h postinfection and were most dramatic at 72 h postinfection. Because upregulation of RPSA and dephosphorylation of cytokeratins 8/18 have been linked with tumor progression, these data suggest that parkin may inhibit cell growth, at least in part, by decreasing RPSA expression and inducing phosphorylation of cytokeratin 8/18.
Collapse
|
15
|
Wang Y, Liu Y, Hu L, Lu F, Jiang Z, Wan C, Wang Z. Laminin receptor 1: a novel protein interacting with human circadian clock protein, hPer1. Neurol Res 2013; 29:429-34. [PMID: 17535553 DOI: 10.1179/016164107x159289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The circadian clock is the central timing system that controls numerous physiologic processes. The current model of these oscillators is based on autoregulatory transcription and translation feedback loops of these circadian genes in which Period1 (Per1) gene occupies a central position. The laminin receptor 1 (Lamr1) and its precursor are expressed in most tissues and play important roles in several physiologic and pathologic processes, including cell differentiation, growth, migration and cancer invasion. The present study showed that Lamr1 was a novel protein that interacted with human circadian clock protein hPer1 by the yeast two-hybrid system and co-immunoprecipitation, which was expressed in many tissues and did not display circadian rhythm. The expression of hPer1 was knocked down to 84.9% by the hPer1 RNA interfering test, but the expression levels of Lamr1 was not depressed by the hPer1 RNA interfering test. The results suggest that Lamr1 is a novel protein that interacts with human circadian clock protein hPer1 and Lamr1 is not a direct efferent element of circadian clock.
Collapse
Affiliation(s)
- Yuhui Wang
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Adhesion and Invasion of Breast and Oesophageal Cancer Cells Are Impeded by Anti-LRP/LR-Specific Antibody IgG1-iS18. PLoS One 2013; 8:e66297. [PMID: 23823499 PMCID: PMC3688881 DOI: 10.1371/journal.pone.0066297] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/03/2013] [Indexed: 12/31/2022] Open
Abstract
Adhesion and invasion have been identified as the two key components of metastasis. The 37 kDa/67 kDa laminin receptor (LRP/LR) is thought to enhance these two processes thus endorsing the progression of cancer. Here we report on LRP/LR and the metastatic potential of MDA-MB 231 breast and WHCO1 oesophageal cancer cells. Western blot analysis revealed a significant increase in total laminin receptor precursor (LRP) levels of breast and oesophageal cancer cells in comparison to non-invasive MCF-7 breast cancer cells, whereas LRP/LR cell surface levels in both cell lines were not significantly different to those of MCF-7 cells as analysed by flow cytometry. Incubation of breast and oesophageal cancer cells with the anti-LRP/LR specific antibody, IgG1-iS18, resulted in significant reduction in the adhesive potential of WHCO1 and MDA-MB 231 cells by 92% and 16%, respectively. Moreover, invasion was significantly impeded by 98% and 25% for WHCO1 and MDA-MB 231 cells, respectively. Pearson's correlation coefficients proved a positive correlation between total LRP/LR levels and invasive potential as well as between the adhesive and invasive potential of breast and oesophageal cancer cells. Our findings suggest that through interference of the LRP/LR-laminin-1 interaction, anti-LRP/LR specific antibody IgG1-iS18 may act as a possible alternative therapeutic tool for metastatic breast and oesophageal cancer treatment.
Collapse
|
17
|
Moodley K, Weiss SFT. Downregulation of the non-integrin laminin receptor reduces cellular viability by inducing apoptosis in lung and cervical cancer cells. PLoS One 2013; 8:e57409. [PMID: 23472084 PMCID: PMC3589420 DOI: 10.1371/journal.pone.0057409] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/21/2013] [Indexed: 12/31/2022] Open
Abstract
The non-integrin laminin receptor, here designated the 37-kDa/67-kDa laminin receptor (LRP/LR), is involved in many physiologically relevant processes, as well as numerous pathological conditions. The overexpression of LRP/LR on various cancerous cell lines plays critical roles in tumour metastasis and angiogenesis. This study investigated whether LRP/LR is implicated in the maintenance of cellular viability in lung and cervical cancer cell lines. Here we show a significant reduction in cellular viability in the aforementioned cell lines as a result of the siRNA-mediated downregulation of LRP. This reduction in cellular viability is due to increased apoptotic processes, reflected by the loss of nuclear integrity and the significant increase in the activity of caspase-3. These results indicate that LRP/LR is involved in the maintenance of cellular viability in tumorigenic lung and cervix uteri cells through the blockage of apoptosis. Knockdown of LRP/LR by siRNA might represent an alternative therapeutic strategy for the treatment of lung and cervical cancer.
Collapse
Affiliation(s)
- Kiashanee Moodley
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Gauteng, The Republic of South Africa
| | - Stefan F. T. Weiss
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Gauteng, The Republic of South Africa
| |
Collapse
|
18
|
Guerra-Rebollo M, Mateo F, Franke K, Huen MS, Lopitz-Otsoa F, Rodríguez MS, Plans V, Thomson TM. Nucleolar exit of RNF8 and BRCA1 in response to DNA damage. Exp Cell Res 2012; 318:2365-76. [DOI: 10.1016/j.yexcr.2012.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 07/05/2012] [Accepted: 07/07/2012] [Indexed: 10/28/2022]
|
19
|
Venticinque L, Meruelo D. Comprehensive proteomic analysis of nonintegrin laminin receptor interacting proteins. J Proteome Res 2012; 11:4863-72. [PMID: 22909348 PMCID: PMC3495180 DOI: 10.1021/pr300307h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Human nonintegrin laminin receptor is a multifunctional protein acting as an integral component of the ribosome and a cell surface receptor for laminin-1. The laminin receptor is overexpressed in several human cancers and is also the cell surface receptor for several viruses and pathogenic prion proteins, making it a pathologically significant protein. This study focused on the proteomic characterization of laminin receptor interacting proteins from Mus musculus. The use of affinity chromatography with immobilized recombinant laminin receptor coupled with mass spectrometry analysis identified 45 proteins with high confidence. Following validation through coimmunoprecipitation, the proteins were classified based on predicted function into ribosomal, RNA processing, signal transduction/metabolism, protein processing, cytoskeleton/cell anchorage, DNA/chromatin, and unknown functions. A significant portion of the identified proteins is related to functions or localizations previously described for laminin receptor. This work represents a comprehensive proteomic approach to studying laminin receptor and provides an essential stepping stone to a better mechanistic understanding of this protein's diverse functions.
Collapse
Affiliation(s)
- Lisa Venticinque
- Gene Therapy Center, Cancer Institute and Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Daniel Meruelo
- Gene Therapy Center, Cancer Institute and Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| |
Collapse
|
20
|
Wang R, Liu X, Küster-Schöck E, Fagotto F. Proteomic analysis of differences in ectoderm and mesoderm membranes by DiGE. J Proteome Res 2012; 11:4575-93. [PMID: 22852788 DOI: 10.1021/pr300379m] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ectoderm and mesoderm can be considered as prototypes for epithelial and mesenchymal cell types. These two embryonic tissues display clear differences in adhesive and motility properties, which are phenomenologically well characterized but remain largely unexplored at the molecular level. Because the key downstream regulations must occur at the plasma membrane and in the underlying actin cortical structures, we have set out to compare the protein content of membrane fractions from Xenopus ectoderm and mesoderm tissues using 2-dimensional difference gel electrophoresis (DiGE). We have thus identified several proteins that are enriched in one or the other tissues, including regulators of the cytoskeleton and of cell signaling. This study represents to our knowledge the first attempt to use proteomics specifically targeted to the membrane-cortex compartment of embryonic tissues. The identified components should help unraveling a variety of tissue-specific functions in the embryo.
Collapse
Affiliation(s)
- Renee Wang
- Department of Biology, McGill University, Montreal, Canada
| | | | | | | |
Collapse
|
21
|
Kim DG, Choi JW, Lee JY, Kim H, Oh YS, Lee JW, Tak YK, Song JM, Razin E, Yun S, Kim S. Interaction of two translational components, lysyl‐tRNA synthetase and p40/37LRP, in plasma membrane promotes laminin‐dependent cell migration. FASEB J 2012; 26:4142-59. [DOI: 10.1096/fj.12-207639] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Dae Gyu Kim
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
- College of PharmacySeoul National UniversitySeoulKorea
| | - Jin Woo Choi
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
- Wellman Center for PhotomedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jin Young Lee
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
- College of PharmacySeoul National UniversitySeoulKorea
| | - Hyerim Kim
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
| | - Young Sun Oh
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
| | - Jung Weon Lee
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
- College of PharmacySeoul National UniversitySeoulKorea
- World Class UniversityDepartment of Molecular Medicine and Biopharmaceutical SciencesSeoul National UniversitySeoulKorea
| | - Yu Kyung Tak
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
- World Class UniversityDepartment of Molecular Medicine and Biopharmaceutical SciencesSeoul National UniversitySeoulKorea
| | - Joon Myong Song
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
- World Class UniversityDepartment of Molecular Medicine and Biopharmaceutical SciencesSeoul National UniversitySeoulKorea
| | - Ehud Razin
- Department of Biochemistry and Molecular BiologyThe Hebrew University‐Hadassah Medical SchoolJerusalemIsrael
| | - Seok‐Hyun Yun
- Wellman Center for PhotomedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Sunghoon Kim
- Medicinal Bioconvergence Research CenterSeoul National UniversitySeoulKorea
- College of PharmacySeoul National UniversitySeoulKorea
- World Class UniversityDepartment of Molecular Medicine and Biopharmaceutical SciencesSeoul National UniversitySeoulKorea
| |
Collapse
|
22
|
Ould-Abeih MB, Petit-Topin I, Zidane N, Baron B, Bedouelle H. Multiple Folding States and Disorder of Ribosomal Protein SA, a Membrane Receptor for Laminin, Anticarcinogens, and Pathogens. Biochemistry 2012; 51:4807-21. [DOI: 10.1021/bi300335r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Mohamed B. Ould-Abeih
- Institut Pasteur, Unit of Molecular Prevention and
Therapy of Human Diseases, Department
of Infection and Epidemiology, rue du Dr. Roux, F-75015 Paris, France
- CNRS, URA3012, rue du Dr. Roux, F-75015 Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur,
rue du Dr. Roux, F-75015 Paris, France
| | - Isabelle Petit-Topin
- Institut Pasteur, Unit of Molecular Prevention and
Therapy of Human Diseases, Department
of Infection and Epidemiology, rue du Dr. Roux, F-75015 Paris, France
- CNRS, URA3012, rue du Dr. Roux, F-75015 Paris, France
| | - Nora Zidane
- Institut Pasteur, Unit of Molecular Prevention and
Therapy of Human Diseases, Department
of Infection and Epidemiology, rue du Dr. Roux, F-75015 Paris, France
- CNRS, URA3012, rue du Dr. Roux, F-75015 Paris, France
| | - Bruno Baron
- Institut Pasteur, Plate-forme
de Biophysique des Macromolécules et de leurs
Interactions, Department of Structural Biology and Chemistry, rue
du Dr. Roux, F-75015 Paris, France
- CNRS, UMR3528, rue du Dr. Roux, 75015
Paris, France
| | - Hugues Bedouelle
- Institut Pasteur, Unit of Molecular Prevention and
Therapy of Human Diseases, Department
of Infection and Epidemiology, rue du Dr. Roux, F-75015 Paris, France
- CNRS, URA3012, rue du Dr. Roux, F-75015 Paris, France
| |
Collapse
|
23
|
Taghian K, Lee JY, Petratos S. Phosphorylation and cleavage of the family of collapsin response mediator proteins may play a central role in neurodegeneration after CNS trauma. J Neurotrauma 2012; 29:1728-35. [PMID: 22181040 DOI: 10.1089/neu.2011.2063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The family of the collapsin response mediator proteins (CRMPs) plays a significant physiological role in neuronal cell bodies and axons within the integrated mammalian central nervous system (CNS). Trauma-induced damage to the CNS results in variable degrees of axonal degeneration, and this may lead to neuronal cell death in key grey matter regions. Site-specific phosphorylation of certain CRMPs has been associated with trauma-induced axonal degeneration. Moreover, recent data implicate the pro-apoptotic, calcium-dependent protease calpain as a key initiator of CRMP cleavage. The primary cleavage product of injury-induced neuronal calpain activation is a C-terminus truncated 55- to 58-kDa form of CRMP, which may exert its effects within the cytoplasm and axonal core, or alternatively through its translocation into the nucleus, initiating neuronal cell death. The precise structure of cleaved CRMP has yet to be elucidated, as is the reason for nuclear translocation. Once the crystal structure of the cytoplasmic and nuclear-translocated forms of CRMPs is determined, a greater molecular understanding of why these forms can initiate neurodegeneration following CNS injury will be established. Such information will be particularly informative in the design of inhibitors of specific protein-protein interaction sites between cleaved CRMP and vital cytosolic or nuclear molecules.
Collapse
Affiliation(s)
- Kasra Taghian
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
24
|
Extraribosomal functions associated with the C terminus of the 37/67 kDa laminin receptor are required for maintaining cell viability. Cell Death Dis 2011; 1:e42. [PMID: 21243100 PMCID: PMC3019570 DOI: 10.1038/cddis.2010.19] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The 37/67 kDa laminin receptor (LAMR) is a multifunctional protein, acting as an extracellular receptor, localizing to the nucleus, and playing roles in rRNA processing and ribosome assembly. LAMR is important for cell viability; however, it is unclear which of its functions are essential. We developed a silent mutant LAMR construct, resistant to siRNA, to rescue the phenotypic effects of knocking down endogenous LAMR, which include inhibition of protein synthesis, cell cycle arrest, and apoptosis. In addition, we generated a C-terminal-truncated silent mutant LAMR construct structurally homologous to the Archaeoglobus fulgidus S2 ribosomal protein and missing the C-terminal 75 residues of LAMR, which displays more sequence divergence. We found that HT1080 cells stably expressing either silent mutant LAMR construct still undergo arrest in the G1 phase of the cell cycle when treated with siRNA. However, the expression of full-length silent mutant LAMR rescues cell viability, whereas the expression of the C-terminal-truncated LAMR does not. Interestingly, we also found that both silent mutant constructs restore protein translation and localize to the nucleus. Our findings indicate that the ability of LAMR to regulate viability is associated with its C-terminal 75 residues. Furthermore, this function is distinct from its role in cell proliferation, independent of its ribosomal functions, and may be regulated by a nonnuclear localization.
Collapse
|
25
|
Venticinque L, Jamieson KV, Meruelo D. Interactions between laminin receptor and the cytoskeleton during translation and cell motility. PLoS One 2011; 6:e15895. [PMID: 21249134 PMCID: PMC3017552 DOI: 10.1371/journal.pone.0015895] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 11/30/2010] [Indexed: 11/19/2022] Open
Abstract
Human laminin receptor acts as both a component of the 40S ribosomal subunit to mediate cellular translation and as a cell surface receptor that interacts with components of the extracellular matrix. Due to its role as the cell surface receptor for several viruses and its overexpression in several types of cancer, laminin receptor is a pathologically significant protein. Previous studies have determined that ribosomes are associated with components of the cytoskeleton, however the specific ribosomal component(s) responsible has not been determined. Our studies show that laminin receptor binds directly to tubulin. Through the use of siRNA and cytoskeletal inhibitors we demonstrate that laminin receptor acts as a tethering protein, holding the ribosome to tubulin, which is integral to cellular translation. Our studies also show that laminin receptor is capable of binding directly to actin. Through the use of siRNA and cytoskeletal inhibitors we have shown that this laminin receptor-actin interaction is critical for cell migration. These data indicate that interactions between laminin receptor and the cytoskeleton are vital in mediating two processes that are intimately linked to cancer, cellular translation and migration.
Collapse
Affiliation(s)
- Lisa Venticinque
- Gene Therapy Center, Cancer Institute and Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Kelly V. Jamieson
- Gene Therapy Center, Cancer Institute and Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Daniel Meruelo
- Gene Therapy Center, Cancer Institute and Department of Pathology, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
26
|
Linge A, Meleady P, Henry M, Clynes M, Kasper M, Barth K. Bleomycin treatment of A549 human lung cancer cells results in association of MGr1-Ag and caveolin-1 in lipid rafts. Int J Biochem Cell Biol 2011; 43:98-105. [DOI: 10.1016/j.biocel.2010.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/09/2010] [Accepted: 10/01/2010] [Indexed: 12/30/2022]
|
27
|
Malygin AA, Babaylova ES, Loktev VB, Karpova GG. A region in the C-terminal domain of ribosomal protein SA required for binding of SA to the human 40S ribosomal subunit. Biochimie 2010; 93:612-7. [PMID: 21167900 DOI: 10.1016/j.biochi.2010.12.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 12/07/2010] [Indexed: 11/25/2022]
Abstract
The human ribosomal protein SA, known also as a precursor of the cell-surface laminin receptor, LAMR, is a protein of the 40S ribosomal subunit. It is homologous to eubacterial ribosomal protein S2p, but has a eukaryote-specific C-terminal domain (CTD) that is responsible in LAMR for the binding of laminin as well as prions and several viruses. Using serial deletions in the SA CTD, we showed that region between amino acids 236-262 is required for binding of the protein to 40S ribosomal subunits. All SA mutants containing this region protected nucleotides in hairpin 40 (which is not bound to any protein in the eubacterial 30S ribosomal subunit) of the 18S rRNA from hydroxyl radical attack. Comparison of our data with the cryo-EM models of the mammalian 40S ribosomal subunit allowed us to locate the SA CTD in the spatial structure of the 40S subunit.
Collapse
Affiliation(s)
- Alexey A Malygin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | | | | |
Collapse
|
28
|
Structure-guided identification of a laminin binding site on the laminin receptor precursor. J Mol Biol 2010; 405:24-32. [PMID: 21040730 DOI: 10.1016/j.jmb.2010.10.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/15/2010] [Accepted: 10/17/2010] [Indexed: 12/19/2022]
Abstract
The 37/ 67-kDa human laminin receptor (LamR) is a cell surface receptor for laminin, prion protein, and a variety of viruses. Because of its wide range of ligands, LamR plays a role in numerous pathologies. LamR overexpression correlates with a highly invasive cell phenotype and increased metastatic ability, mediated by interactions between LamR and laminin. In addition, the specific targeting of LamR with small interfering RNAs, blocking antibodies, and Sindbis viral vectors confers anti-tumor effects. We adopted a structure-based approach to map a laminin binding site on human LamR by comparing the sequences and crystal structures of LamR and Archaeoglobus fulgidus S2p, a non-laminin-binding ortholog. Here, we identify a laminin binding site on LamR, comprising residues Phe32, Glu35, and Arg155, which are conserved among mammalian species. Mutation of these residues results in a significant loss of laminin binding. Further, recombinant wild-type LamR is able to act as a soluble decoy to inhibit cellular migration towards laminin. Mutation of this laminin binding site results in loss of migration inhibition, which demonstrates the physiological role of Phe32, Glu35, and Arg155 for laminin binding activity. Mapping of the LamR binding site should contribute to the development of therapeutics that inhibit LamR interactions with laminin and may aid in the prevention of tumor growth and metastasis.
Collapse
|
29
|
Mathew S, Fu L, Hasebe T, Ishizuya-Oka A, Shi YB. Tissue-dependent induction of apoptosis by matrix metalloproteinase stromelysin-3 during amphibian metamorphosis. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2010; 90:55-66. [PMID: 20301218 PMCID: PMC3412310 DOI: 10.1002/bdrc.20170] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Matrix metalloproteinases (MMPs) are a superfamily of Zn(2+)-dependent proteases that are capable of cleaving the proteinaceous component of the extracellular matrix (ECM). The ECM is a critical medium for cell-cell interactions and can also directly signal cells through cell surface ECM receptors, such as integrins. In addition, many growth factors and signaling molecules are stored in the ECM. Thus, ECM remodeling and/or degradation by MMPs are expected to affect cell fate and behavior during many developmental and pathological processes. Numerous studies have shown that the expression of MMP mRNAs and proteins associates tightly with diverse developmental and pathological processes, such as tumor metastasis and mammary gland involution. In vivo evidence to support the roles of MMPs in these processes has been much harder to get. Here, we will review some of our studies on MMP11, or stromelysin-3, during the thyroid hormone-dependent amphibian metamorphosis, a process that resembles the so-called postembryonic development in mammals (from a few months before to several months after birth in humans when organ growth and maturation take place). Our investigations demonstrate that stromelysin-3 controls apoptosis in different tissues via at least two distinct mechanisms.
Collapse
Affiliation(s)
- Smita Mathew
- Section on Molecular Morphogenesis, Laboratory of Gene Regulation and Development, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, 20892
| | - Liezhen Fu
- Section on Molecular Morphogenesis, Laboratory of Gene Regulation and Development, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, 20892
| | - Takashi Hasebe
- Department of Biology, Nippon Medical School, Kawasaki, Kanagawa 211-0063, Japan
| | - Atsuko Ishizuya-Oka
- Department of Biology, Nippon Medical School, Kawasaki, Kanagawa 211-0063, Japan
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Laboratory of Gene Regulation and Development, Program in Cellular Regulation and Metabolism (PCRM), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, 20892
| |
Collapse
|
30
|
Malygin AA, Bondarenko EI, Ivanisenko VA, Protopopova EV, Karpova GG, Loktev VB. C-terminal fragment of human laminin-binding protein contains a receptor domain for Venezuelan equine encephalitis and tick-borne encephalitis viruses. BIOCHEMISTRY (MOSCOW) 2010; 74:1328-36. [DOI: 10.1134/s0006297909120050] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
FAAP is Involved in Cell Attachment*. PROG BIOCHEM BIOPHYS 2009. [DOI: 10.3724/sp.j.1206.2008.00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Malygin AA, Bochkaeva ZV, Bondarenko EI, Kossinova OA, Loktev VB, Shatsky IN, Karpova GG. Binding of the IRES of hepatitis C virus RNA to the 40S ribosomal subunit: Role of p40. Mol Biol 2009. [DOI: 10.1134/s0026893309060120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
33
|
Liu W, Zhou XW, Liu S, Hu K, Wang C, He Q, Li M. Calpain-truncated CRMP-3 and -4 contribute to potassium deprivation-induced apoptosis of cerebellar granule neurons. Proteomics 2009; 9:3712-28. [PMID: 19639589 DOI: 10.1002/pmic.200800979] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Increasing evidence shows that calpain-mediated proteolytic processing of a selective number of proteins plays an important role in neuronal apoptosis. Study of calpain-mediated cleavage events and related functions may contribute to a better understanding of neuronal apoptosis and neurodegenerative diseases. We, therefore, investigated the role of calpain substrates in potassium deprivation-induced apoptosis of cerebellar granule neurons (CGNs). Twelve previously known and seven novel candidates of calpain substrates were identified by 2-D DIGE and MALDI-TOF/TOF MS analysis. Further, the identified novel calpain substrates were validated by Western blot analysis. Moreover, we focused on the collapsin response mediator proteins (CRMP-1, -2, -3 and -4 isoforms) and found that CRMPs were proteolytically processed by calpain but not by caspase, both in vivo and in vitro. To clarify the properties of the calpain-mediated proteolysis of CRMPs, we constructed the deletion mutants of CRMPs for additional biochemical studies. In vitro cleavage assays revealed that CRMP-1, -2 and -4 were truncated by calpain at the C-terminus, whereas CRMP-3 was cleaved at the N-terminus. Finally, we assessed the role of CRMPs in the process of potassium deprivation-triggered neuronal apoptosis by overexpressing the truncated CRMPs in CGNs. Our data clearly showed that the truncated CRMP-3 and -4, but not CRMP-1 and -2, significantly induced neuronal apoptosis. These findings demonstrated that calpain-truncated CRMP-3 and -4 act as pro-apoptotic players when CGNs undergo apoptosis.
Collapse
Affiliation(s)
- Wei Liu
- Proteomics Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, PR China
| | | | | | | | | | | | | |
Collapse
|
34
|
Multiple functions of the 37/67-kd laminin receptor make it a suitable target for novel cancer gene therapy. Mol Ther 2009; 18:63-74. [PMID: 19724263 DOI: 10.1038/mt.2009.199] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The 37/67-kd laminin receptor, LAMR, is a multifunctional protein that associates with the 40S ribosomal subunit and also localizes to the cell membrane to interact with the extracellular matrix. LAMR is overexpressed in many types of cancer, playing important roles in tumor-cell migration and invasion. Here, we show that LAMR is also vital for tumor-cell proliferation, survival, and protein translation. Small-interfering RNA (siRNA)-mediated reduction in expression of LAMR leads to G1 phase cell-cycle arrest in vitro by altering cyclins A2/B1, cyclin-dependent kinases (CDKs) 1/2, Survivin, and p21 expression levels. In vivo, reduction in LAMR expression results in inhibition of HT1080 cells to develop tumors. We also found that LAMR's ribosomal functions are critical for translation as reduction in LAMR expression leads to a dramatic decrease in newly synthesized proteins. Further, cells with lower expression of LAMR have fewer 40S subunits and 80S monosomes, causing an increase in free 60S ribosomal subunits. These results indicate that LAMR is able to regulate tumor development in many ways; further enhancing its potential as a target for gene therapy. To test this, we developed a novel Sindbis/Lenti pseudotype vector carrying short-hairpin RNA (shRNA) designed against lamr. This pseudotype vector effectively reduces LAMR expression and specifically targets tumors in vivo. Treatment of tumor-bearing severe combine immunodeficient (SCID) mice with this pseudotype vector significantly inhibits tumor growth. Thus, we show that LAMR can be used as a target in novel therapy for tumor reduction and elimination.
Collapse
|
35
|
Pflanz H, Vana K, Mitteregger G, Pace C, Messow D, Sedlaczek C, Nikles D, Kretzschmar HA, Weiss SFT. Microinjection of lentiviral vectors expressing small interfering RNAs directed against laminin receptor precursor mRNA prolongs the pre-clinical phase in scrapie-infected mice. J Gen Virol 2009; 90:269-74. [PMID: 19088298 DOI: 10.1099/vir.0.004168-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We examined therapeutic in vitro and in vivo approaches using lentivirus-based packaging of small interfering RNAs (siRNAs) targeting the non-integrin laminin receptor mRNA for treatment and prevention of prion disorders. Transfection of N2aSc(+) cells with recombinant plasmids expressing three different siRNAs, significantly reduced both the LRP (laminin receptor precursor) and PrP(Sc) levels by approximately 40-60 %. Stereotactic intracerebral microinjection of recombinant lentiviral vectors LVsiRNA-LRP 7 and 9 into the cortex of C57BL/6 wild-type mice resulted in a significant reduction of the LR levels in the cortex 15 days post-injection by 62 and 82 %, respectively. Intracerebral RML inoculation of C57BL/6 mice after microinjection with recombinant lentiviral vector LVsiRNA-LRP 7 into the hippocampus resulted in a significant reduction of both LRP and PrP(Sc) levels by 36 and 41 %, respectively, concomitant with a significant prolongation of the pre-clinical phase. Lentiviral vectors expressing siRNAs targeting LRP mRNA represent a novel delivery system for the treatment of transmissible spongiform encephalopathies.
Collapse
Affiliation(s)
- Heike Pflanz
- Laboratorium für Molekulare Biologie - Genzentrum - Institut für Biochemie der LMU München, Feodor-Lynen Str. 25, D-81377 München, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kossinova OA, Malygin AA, Babailova ES, Karpova GG. Binding of human ribosomal protein p40 and its truncated mutants to the small ribosomal subunit. Mol Biol 2008. [DOI: 10.1134/s0026893308060125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Qiu J, Choi G, Li L, Wang H, Pitteri SJ, Pereira-Faca SR, Krasnoselsky AL, Randolph TW, Omenn GS, Edelstein C, Barnett MJ, Thornquist MD, Goodman GE, Brenner DE, Feng Z, Hanash SM. Occurrence of autoantibodies to annexin I, 14-3-3 theta and LAMR1 in prediagnostic lung cancer sera. J Clin Oncol 2008; 26:5060-6. [PMID: 18794547 DOI: 10.1200/jco.2008.16.2388] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE We have implemented a high throughput platform for quantitative analysis of serum autoantibodies, which we have applied to lung cancer for discovery of novel antigens and for validation in prediagnostic sera of autoantibodies to antigens previously defined based on analysis of sera collected at the time of diagnosis. MATERIALS AND METHODS Proteins from human lung adenocarcinoma cell line A549 lysates were subjected to extensive fractionation. The resulting 1,824 fractions were spotted in duplicate on nitrocellulose-coated slides. The microarrays produced were used in a blinded validation study to determine whether annexin I, PGP9.5, and 14-3-3 theta antigens previously found to be targets of autoantibodies in newly diagnosed patients with lung cancer are associated with autoantibodies in sera collected at the presymptomatic stage and to determine whether additional antigens may be identified in prediagnostic sera. Individual sera collected from 85 patients within 1 year before a diagnosis of lung cancer and 85 matched controls from the Carotene and Retinol Efficacy Trial (CARET) cohort were hybridized to individual microarrays. RESULTS We present evidence for the occurrence in lung cancer sera of autoantibodies to annexin I, 14-3-3 theta, and a novel lung cancer antigen, LAMR1, which precede onset of symptoms and diagnosis. CONCLUSION Our findings suggest potential utility of an approach to diagnosis of lung cancer before onset of symptoms that includes screening for autoantibodies to defined antigens.
Collapse
Affiliation(s)
- Ji Qiu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The 67LR (67 kDa laminin receptor) is a cell-surface receptor with high affinity for its primary ligand. Its role as a laminin receptor makes it an important molecule both in cell adhesion to the basement membrane and in signalling transduction following this binding event. The protein also plays critical roles in the metastasis of tumour cells. Isolation of the protein from either normal or cancerous cells results in a product with an approx. molecular mass of 67 kDa. This protein is believed to be derived from a smaller precursor, the 37LRP (37 kDa laminin receptor precursor). However, the precise mechanism by which cytoplasmic 37LRP becomes cell-membrane-embedded 67LR is unclear. The process may involve post-translational fatty acylation of the protein combined with either homo- or hetero-dimerization, possibly with a galectin-3-epitope-containing partner. Furthermore, it has become clear that acting as a receptor for laminin is not the only function of this protein. 67LR also acts as a receptor for viruses, such as Sindbis virus and dengue virus, and is involved with internalization of the prion protein. Interestingly, unmodified 37LRP is a ribosomal component and homologues of this protein are found in all five kingdoms. In addition, it appears to be strongly associated with histones in the eukaryotic cell nucleus, although the precise role of these interactions is not clear. Here we review the current understanding of the structure and function of this molecule, as well as highlighting areas requiring further research.
Collapse
|
39
|
Zuber C, Knackmuss S, Zemora G, Reusch U, Vlasova E, Diehl D, Mick V, Hoffmann K, Nikles D, Fröhlich T, Arnold GJ, Brenig B, Wolf E, Lahm H, Little M, Weiss S. Invasion of Tumorigenic HT1080 Cells Is Impeded by Blocking or Downregulating the 37-kDa/67-kDa Laminin Receptor. J Mol Biol 2008; 378:530-9. [DOI: 10.1016/j.jmb.2008.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Revised: 01/25/2008] [Accepted: 02/01/2008] [Indexed: 11/27/2022]
|
40
|
siRNA-mediated silencing of the 37/67-kDa high affinity laminin receptor in Hep3B cells induces apoptosis. Cell Mol Biol Lett 2008; 13:452-64. [PMID: 18425431 PMCID: PMC6275979 DOI: 10.2478/s11658-008-0017-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Accepted: 01/25/2008] [Indexed: 12/12/2022] Open
Abstract
The laminin-binding protein, variously called the 37/67-kDa high affinity laminin receptor or p40, mediates the attachment of normal cells to the laminin network, and also has a role as a ribosomal protein. Over-expression of this protein has been strongly correlated with the metastatic phenotype. However, few studies have investigated the cellular consequence of the ablation of this gene’s expression. To address this issue, the expression of the 37/67-kDa high affinity laminin receptor was knocked out with several siRNA constructs via RNA interference in transformed liver (Hep3B) cells. In each case where the message was specifically ablated, apoptosis was induced, as determined by annexin V/propidium iodide staining, and by double staining with annexin V and an antibody directed against the 37/67-kDa high affinity laminin receptor. These results suggest that this protein plays a critical role in maintaining cell viability.
Collapse
|
41
|
Marcos-Carcavilla A, Calvo JH, González C, Serrano C, Moazami-Goudarzi K, Laurent P, Bertaud M, Hayes H, Beattie AE, Lyahyai J, Martín-Burriel I, Torres JM, Serrano M. Structural and functional analysis of the ovine laminin receptor gene (RPSA): Possible involvement of the LRP/LR protein in scrapie response. Mamm Genome 2008; 19:92-105. [PMID: 18202837 DOI: 10.1007/s00335-007-9085-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 11/23/2007] [Indexed: 11/28/2022]
Abstract
Scrapie is a prion disease affecting sheep and goats. Susceptibility to this neurodegenerative disease shows polygenic variance. The involvement of the laminin receptor (LRP/LR) in the metabolism and propagation of prions has previously been demonstrated. In the present work, the ovine laminin receptor gene (RPSA) was isolated, characterized, and mapped to ovine chromosome OAR19q13. Real-time RT-PCR revealed a significant decrease in RPSA mRNA in cerebellum after scrapie infection. Conversely, no differences were detected in other brain regions such as diencephalon and medulla oblongata. Association analysis showed that a polymorphism reflecting the presence of a RPSA pseudogene was overrepresented in a group of sheep resistant to scrapie infection. No amino acid change in the LRP/LR protein was found in the 126 sheep analyzed. However, interesting amino acid positions (241, 272, and 290), which could participate in the species barrier to scrapie and maybe to other transmissible spongiform encephalopathies, were identified by comparing LRP/LR sequences from various mammals with variable levels of resistance to scrapie.
Collapse
Affiliation(s)
- Ane Marcos-Carcavilla
- Departamento de Mejora Genética Animal, INIA, Ctra La Coruña Km 7.5, Madrid, 28040, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Jamieson KV, Wu J, Hubbard SR, Meruelo D. Crystal structure of the human laminin receptor precursor. J Biol Chem 2007; 283:3002-3005. [PMID: 18063583 DOI: 10.1074/jbc.c700206200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human laminin receptor (LamR) interacts with many ligands, including laminin, prions, Sindbis virus, and the polyphenol (-)-epigallocatechin-3-gallate (EGCG), and has been implicated in a number of diseases. LamR is overexpressed on tumor cells, and targeting LamR elicits anti-cancer effects. Here, we report the crystal structure of human LamR, which provides insights into its function and should facilitate the design of novel therapeutics targeting LamR.
Collapse
Affiliation(s)
- Kelly V Jamieson
- Gene Therapy Center, Cancer Institute and Department of Pathology, New York University School of Medicine, New York, New York 10016
| | - Jinhua Wu
- Structural Biology Program, Kimmel Center for Biology and Medicine of the Skirball Institute, and Department of Pharmacology, New York University School of Medicine, New York, New York 10016
| | - Stevan R Hubbard
- Structural Biology Program, Kimmel Center for Biology and Medicine of the Skirball Institute, and Department of Pharmacology, New York University School of Medicine, New York, New York 10016.
| | - Daniel Meruelo
- Gene Therapy Center, Cancer Institute and Department of Pathology, New York University School of Medicine, New York, New York 10016.
| |
Collapse
|
43
|
Shi YB, Fu L, Hasebe T, Ishizuya-Oka A. Regulation of extracellular matrix remodeling and cell fate determination by matrix metalloproteinase stromelysin-3 during thyroid hormone-dependent post-embryonic development. Pharmacol Ther 2007; 116:391-400. [PMID: 17919732 DOI: 10.1016/j.pharmthera.2007.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 07/30/2007] [Indexed: 01/23/2023]
Abstract
Interactions between cells and extracellular matrix (ECM), in particular the basement membrane (BM), are fundamentally important for the regulation of a wide variety of physiological and pathological processes. Matrix metalloproteinases (MMP) play critical roles in ECM remodeling and/or regulation of cell-ECM interactions because of their ability to cleave protein components of the ECM. Of particular interest among MMP is stromelysin-3 (ST3), which was first isolated from a human breast cancer and also shown to be correlated with apoptosis during development and invasion of tumor cells in mammals. We have been using intestinal remodeling during thyroid hormone (TH)-dependent amphibian metamorphosis as a model to study the role of ST3 during post-embryonic tissue remodeling and organ development in vertebrates. This process involves complete degeneration of the tadpole or larval epithelium through apoptosis and de novo development of the adult epithelium. Here, we will first summarize expression studies by us and others showing a tight spatial and temporal correlation of the expression of ST3 mRNA and protein with larval cell death and adult tissue development. We will then review in vitro and in vivo data supporting a critical role of ST3 in TH-induced larval epithelial cell death and ECM remodeling. We will further discuss the potential mechanisms of ST3 function during metamorphosis and its broader implications.
Collapse
Affiliation(s)
- Yun-Bo Shi
- Laboratory of Gene Regulation and Development, National Institute of Child Health and Human Development, National Institutes of Health, Building 18T, Room 106, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
44
|
Nam HJ, Lane MD, Padron E, Gurda B, McKenna R, Kohlbrenner E, Aslanidi G, Byrne B, Muzyczka N, Zolotukhin S, Agbandje-McKenna M. Structure of adeno-associated virus serotype 8, a gene therapy vector. J Virol 2007; 81:12260-71. [PMID: 17728238 PMCID: PMC2168965 DOI: 10.1128/jvi.01304-07] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adeno-associated viruses (AAVs) are being developed as gene therapy vectors, and their efficacy could be improved by a detailed understanding of their viral capsid structures. AAV serotype 8 (AAV8) shows a significantly greater liver transduction efficiency than those of other serotypes, which has resulted in efforts to develop this virus as a gene therapy vector for hemophilia A and familial hypercholesterolemia. Pseudotyping studies show that the differential tissue tropism and transduction efficiencies exhibited by the AAVs result from differences in their capsid viral protein (VP) amino acids. Towards identifying the structural features underpinning these disparities, we report the crystal structure of the AAV8 viral capsid determined to 2.6-A resolution. The overall topology of its common overlapping VP is similar to that previously reported for the crystal structures of AAV2 and AAV4, with an eight-stranded beta-barrel and long loops between the beta-strands. The most significant structural differences between AAV8 and AAV2 (the best-characterized serotype) are located on the capsid surface at protrusions surrounding the two-, three-, and fivefold axes at residues reported to control transduction efficiency and antibody recognition for AAV2. In addition, a comparison of the AAV8 and AAV2 capsid surface amino acids showed a reduced distribution of basic charge for AAV8 at the mapped AAV2 heparin sulfate receptor binding region, consistent with an observed non-heparin-binding phenotype for AAV8. Thus, this AAV8 structure provides an additional platform for mutagenesis efforts to characterize AAV capsid regions responsible for differential cellular tropism, transduction, and antigenicity for these promising gene therapy vectors.
Collapse
Affiliation(s)
- Hyun-Joo Nam
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Amano T, Fu L, Marshak A, Kwak O, Shi YB. Spatio-temporal regulation and cleavage by matrix metalloproteinase stromelysin-3 implicate a role for laminin receptor in intestinal remodeling during Xenopus laevis metamorphosis. Dev Dyn 2007; 234:190-200. [PMID: 16059908 DOI: 10.1002/dvdy.20511] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The 37-kd laminin receptor precursor (LR) was first identified as a 67-kd protein that binds laminin with high affinity. We have recently isolated the Xenopus laevis LR as an in vitro substrate of matrix metalloproteinase stromelysin-3 (ST3), which is highly upregulated during intestinal metamorphosis in Xenopus laevis. Here, we show that LR is expressed in the intestinal epithelium of premetamorphic tadpoles. During intestinal metamorphosis, LR is downregulated in the apoptotic epithelium and concurrently upregulated in the connective tissue but with little expression in the developing adult epithelium. Toward the end of metamorphosis, as adult epithelial cells differentiate, they begin to express LR. Furthermore, LR is cleaved during intestinal remodeling when ST3 is highly expressed or in premetamorphic intestine of transgenic tadpoles overexpressing ST3. These results suggest that LR plays a role in cell fate determination and tissue morphogenesis, in part through its cleavage by ST3. Interestingly, high levels of LR are known to be expressed in tumor cells, which are often surrounded by fibroblasts expressing ST3, suggesting that LR cleavage by ST3 plays a role in both physiological and pathological processes.
Collapse
Affiliation(s)
- Tosikazu Amano
- Laboratory of Gene Regulation and Development, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
46
|
Pluder F, Sinz A, Beck-Sickinger AG. Proliferative effect of calcitonin gene-related peptide is induced by at least five proteins as identified by proteome profiling. Chem Biol Drug Des 2007; 69:14-22. [PMID: 17313453 DOI: 10.1111/j.1747-0285.2007.00466.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Calcitonin gene-related peptide is a 37 amino acid neuropeptide. Although calcitonin gene-related peptide activates a G-protein-coupled receptor, recent evidence suggests that calcitonin gene-related peptide induces more complex signaling cascades including the activation of MAP kinases [Eur J Pharmacol; 389:125-130 (2000), Neuropeptides; 34:229-233 (2000)]. However, the molecular mechanisms of this activation still remain to be elucidated. For the first time we applied a proteomics approach in order to identify molecular targets of calcitonin gene-related peptide downstream signaling in the neuroblastoma cell line SK-N-MC and identified proteins that changed either their expression, location, or their post-translational modifications in a time-dependent manner after calcitonin gene-related peptide stimulation.
Collapse
Affiliation(s)
- Franka Pluder
- Institute of Biochemistry, Faculty of Bioscience, Pharmacy and Psychology, University of Leipzig, Brüderstrasse 34, D-04103 Leipzig, Germany
| | | | | |
Collapse
|
47
|
Vana K, Zuber C, Nikles D, Weiss S. Novel aspects of prions, their receptor molecules, and innovative approaches for TSE therapy. Cell Mol Neurobiol 2007; 27:107-28. [PMID: 17151946 DOI: 10.1007/s10571-006-9121-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 09/20/2006] [Indexed: 10/23/2022]
Abstract
1. Prion diseases are a group of rare, fatal neurodegenerative diseases, also known as transmissible spongiform encephalopathies (TSEs), that affect both animals and humans and include bovine spongiform encephalopathy (BSE) in cattle, scrapie in sheep, chronic wasting disease (CWD) in deer and elk, and Creutzfeldt-Jakob disease (CJD) in humans. TSEs are usually rapidly progressive and clinical symptoms comprise dementia and loss of movement coordination due to the accumulation of an abnormal isoform (PrP(Sc)) of the host-encoded prion protein (PrP(c)). 2. This article reviews the current knowledge on PrP(c) and PrP(Sc), prion replication mechanisms, interaction partners of prions, and their cell surface receptors. Several strategies, summarized in this article, have been investigated for an effective antiprion treatment including development of a vaccination therapy and screening for potent chemical compounds. Currently, no effective treatment for prion diseases is available. 3. The identification of the 37 kDa/67 kDa laminin receptor (LRP/LR) and heparan sulfate as cell surface receptors for prions, however, opens new avenues for the development of alternative TSE therapies.
Collapse
Affiliation(s)
- Karen Vana
- Laboratorium für Molekulare Biologie, Genzentrum, Institut für Biochemie der Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, München, Germany
| | | | | | | |
Collapse
|
48
|
Senapin S, Phongdara A. Binding of shrimp cellular proteins to Taura syndrome viral capsid proteins VP1, VP2 and VP3. Virus Res 2006; 122:69-77. [PMID: 16889862 DOI: 10.1016/j.virusres.2006.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2006] [Revised: 06/23/2006] [Accepted: 06/28/2006] [Indexed: 11/21/2022]
Abstract
Viruses are a major cause of production losses in the world shrimp-farming industry. Despite this, little is known about viral-host interactions in shrimp due in part to the lack of continuous shrimp cell lines. Here, the yeast two-hybrid assay system was employed to study interactions between three Taura syndrome viral capsid proteins (VP1-VP3) and proteins from a cDNA library of the black tiger shrimp Penaeus monodon. VP1 interacted with beta-actin, elongation factor 1alpha (EF1alpha), lysozyme (Lys) and laminin receptor/ribosomal protein p40 (Lamr/p40) containing a putative palindromic laminin binding region LMWWML. VP2 interacted with beta-actin and EF1alpha, while VP3 bound to the same proteins as VP1 except for Lamr/p40. In vitro pull-down assays confirmed these interactions. The most interesting interaction was specific binding between VP1 and Lamr/p40 since Lamr/p40 has been identified as the mammalian cell receptor for several arthropod-borne viruses (arboviruses). A search of mosquito vector and Drosophila sequences at available databases revealed the presence of putative Lamr/p40 proteins with high homology to the Lamr/p40 from shrimp.
Collapse
Affiliation(s)
- Saengchan Senapin
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand.
| | | |
Collapse
|
49
|
Amano T, Kwak O, Fu L, Marshak A, Shi YB. The matrix metalloproteinase stromelysin-3 cleaves laminin receptor at two distinct sites between the transmembrane domain and laminin binding sequence within the extracellular domain. Cell Res 2005; 15:150-9. [PMID: 15780176 DOI: 10.1038/sj.cr.7290280] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The matrix metalloproteinase (MMP) stromelysin-3 (ST3) has long been implicated to play an important role in extracellular matrix (ECM) remodeling and cell fate determination during normal and pathological processes. However, like other MMPs, the molecular basis of ST3 function in vivo remains unclear due to the lack of information on its physiological substrates. Furthermore, ST3 has only weak activities toward all tested ECM proteins. Using thyroid hormone-dependent Xenopus laevis metamorphosis as a model, we demonstrated previously that ST3 is important for apoptosis and tissue morphogenesis during intestinal remodeling. Here, we used yeast two-hybrid screen with mRNAs from metamorphosing tadpoles to identify potential substrate of ST3 during development. We thus isolated the 37 kd laminin receptor precursor (LR). We showed that LR binds to ST3 in vitro and can be cleaved by ST3 at two sites, distinct from where other MMPs cleave. Through peptide sequencing, we determined that the two cleavage sites are in the extracellular domain between the transmembrane domain and laminin binding sequence. Furthermore, we demonstrated that these cleavage sites are conserved in human LR. These results together with high levels of human LR and ST3 expression in carcinomas suggest that LR is a likely in vivo substrate of ST3 and that its cleavage by ST3 may alter cell-extracellular matrix interaction, thus, playing a role in mediating the effects of ST3 on cell fate and behavior observed during development and pathogenesis.
Collapse
Affiliation(s)
- Tosikazu Amano
- Laboratory of Gene Regulation and Development, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
50
|
Sun L, Shi Y, Guo C, Yao L, Lin T, Du J, Han Q, Han Y, Fan D. Regulation of multidrug resistance by MGr1-antigen in gastric cancer cells. Tumour Biol 2005; 27:27-35. [PMID: 16340247 DOI: 10.1159/000090153] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2005] [Accepted: 06/21/2005] [Indexed: 11/19/2022] Open
Abstract
Previously, a novel protein, MGr1-Ag, was associated with tumor multidrug resistance (MDR), and the role and the underlying mechanisms of MGr1-Ag in MDR of gastric cancer cells were characterized. Initial studies using the introduction of sense or antisense vectors for MGr1-Ag resulted in the genetical up- or downregulation of MGr1-Ag in gastric cancer cells, respectively. Subsequent studies revealed the expression of MGr1-Ag, P-glycoprotein (P-gp), MDR-associated protein (MRP), Bcl-2 and Bax in gastric cancer cells via Western blot analysis. The sensitivity of gastric cancer cells to chemotherapeutic drugs was assessed using the colony-forming assay, and Adriamycin (ADM) accumulation was evaluated by flow cytometry. Further study of ADM-induced apoptosis was detected by annexin-V/propidium iodide staining. The expression level of MGr1-Ag in MDR gastric cancer cells is much higher than that in their parental cells. Overexpression of exogenous MGr1-Ag may promote the MDR phenotype of gastric cancer cells, decrease intracellular ADM accumulation and protect gastric cancer cells from ADM-induced apoptosis, whereas downregulation of MGr1-Ag had reverse effects. Western blot analysis suggested that MGr1-Ag may regulate the expression of P-gp, MRP, Bcl-2 and Bax. In conclusion, MGr1-Ag may promote MDR of gastric cancer cells via a decrease in intracellular drug accumulation and inhibition of drug-induced apoptosis.
Collapse
Affiliation(s)
- Li Sun
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|