1
|
Hu T, Zhang Y, Yang T, He Q, Zhao M. LYPD3, a New Biomarker and Therapeutic Target for Acute Myelogenous Leukemia. Front Genet 2022; 13:795820. [PMID: 35360840 PMCID: PMC8963240 DOI: 10.3389/fgene.2022.795820] [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: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Acute myelogenous leukemia (AML) is nosocomial with the highest pediatric mortality rates and a relatively poor prognosis. C4.4A(LYPD3) is a tumorigenic and high-glycosylated cell surface protein that has been proven to be linked with the carcinogenic effects in solid tumors, but no hematologic tumors have been reported. We focus on exploring the molecular mechanism of LYPD3 in the regulation of the occurrence and development of AML to provide a research basis for the screening of markers related to the treatment and prognosis. Methods: Datasets on RNA Sequencing (RNA-seq) and mRNA expression profiles of 510 samples were obtained from The Cancer Genome Atlas Program/The Genotype-Tissue Expression (Tcga-gtex) on 10 March 2021, which included the information on 173 AML tumorous tissue samples and 337 normal blood samples. The differential expression, identification of prognostic genes based on the COX regression model, and LASSO regression were analyzed. In order to better verify, experiments including gene knockdown mediated by small interfering RNA (siRNA), cell proliferation assays, and Western blot were prefomed. We studied the possible associated pathways through which LYPD3 may have an impact on the pathogenesis and prognosis of AML by gene set enrichment analysis (GSEA). Results: A total of 11,490 differential expression genes (DEGs) were identified. Among them, 4,164 genes were upregulated, and 7,756 genes were downregulated. The univariate Cox regression analysis and LASSO regression analysis found that 28 genes including LYPD3, DNAJC8, and other genes were associated with overall survival (OS). After multivariate Cox analysis, a total of 10 genes were considered significantly correlated with OS in AML including LYPD3, which had a poor impact on AML (p <0.05). The experiment results also supported the above conclusion. We identified 25 pathways, including the E2F signaling pathway, p53 signaling pathway, and PI3K_AKT signaling pathway, that were significantly upregulated in AML samples with high LYPD3 expression (p < 0.05) by GSEA. Further, the results of the experiment suggested that LYPD3 participates in the development of AML through the p53 signaling pathway or/and PI3K/AKT signaling pathway. Conclusion: This study first proved that the expression of LYPD3 was elevated in AML, which was correlated with poor clinical characteristics and prognosis. In addition, LYPD3 participates in the development of AML through p53 or/and the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yingjie Zhang
- College of Biology, Hunan University, Changsha, China
| | - Tianqing Yang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingnan He
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Qingnan He, ; Mingyi Zhao,
| | - Mingyi Zhao
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Qingnan He, ; Mingyi Zhao,
| |
Collapse
|
2
|
Jiang Y, Lin L, Chen S, Jiang L, Kriegbaum MC, Gårdsvoll H, Hansen LV, Li J, Ploug M, Yuan C, Huang M. Crystal Structures of Human C4.4A Reveal the Unique Association of Ly6/uPAR/α-neurotoxin Domain. Int J Biol Sci 2020; 16:981-993. [PMID: 32140067 PMCID: PMC7053344 DOI: 10.7150/ijbs.39919] [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: 09/02/2019] [Accepted: 12/26/2019] [Indexed: 01/26/2023] Open
Abstract
Ly6/uPAR/α-neurotoxin domain (LU-domain) is characterized by the presence of 4-5 disulfide bonds and three flexible loops that extend from a core stacked by several conversed disulfide bonds (thus also named three-fingered protein domain). This highly structurally stable protein domain is typically a protein-binder at extracellular space. Most LU proteins contain only single LU-domain as represented by Ly6 proteins in immunology and α-neurotoxins in snake venom. For Ly6 proteins, many are expressed in specific cell lineages and in differentiation stages, and are used as markers. In this study, we report the crystal structures of the two LU-domains of human C4.4A alone and its complex with a Fab fragment of a monoclonal anti-C4.4A antibody. Interestingly, both structures showed that C4.4A forms a very compact globule with two LU-domain packed face to face. This is in contrast to the flexible nature of most LU-domain-containing proteins in mammals. The Fab combining site of C4.4A involves both LU-domains, and appears to be the binding site for AGR2, a reported ligand of C4.4A. This work reports the first structure that contain two LU-domains and provides insights on how LU-domains fold into a compact protein and interacts with ligands.
Collapse
Affiliation(s)
- Yunbin Jiang
- State Key Laboratory of Structure Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Lin
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China
| | - Shanli Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Mette C Kriegbaum
- Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen N, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2220 Copenhagen N, Denmark
| | - Henrik Gårdsvoll
- Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen N, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2220 Copenhagen N, Denmark
| | - Line V Hansen
- Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen N, Denmark
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Michael Ploug
- Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen N, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2220 Copenhagen N, Denmark
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Mingdong Huang
- State Key Laboratory of Structure Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China.,College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Lin Q, Huang Y, Giordano FJ, Yun Z. Generation of a hypoxia-sensing mouse model. Genesis 2019; 58:e23352. [PMID: 31872977 DOI: 10.1002/dvg.23352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 11/08/2022]
Abstract
Oxygen (O2 ) homeostasis is essential to the metazoan life. O2 -sensing or hypoxia-regulated molecular pathways are intimately involved in a wide range of critical cellular functions and cell survival from embryogenesis to adulthood. In this report, we have designed an innovative hypoxia sensor (O2 CreER) based on the O2 -dependent degradation domain of the hypoxia-inducible factor-1α and Cre recombinase. We have further generated a hypoxia-sensing mouse model, R26-O2 CreER, by targeted insertion of the O2 CreER-coding cassette in the ROSA26 locus. Using the ROSAmTmG mouse strain as a reporter, we have found that this novel hypoxia-sensing mouse model can specifically identify hypoxic cells under the pathological condition of hind-limb ischemia in adult mice. This model can also label embryonic cells including vibrissal follicle cells in E13.5-E15.5 embryos. This novel mouse model offers a valuable genetic tool for the study of hypoxia and O2 sensing in mammalian systems under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Qun Lin
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| | - Yan Huang
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Frank J Giordano
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Zhong Yun
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
4
|
Leth JM, Leth-Espensen KZ, Kristensen KK, Kumari A, Lund Winther AM, Young SG, Ploug M. Evolution and Medical Significance of LU Domain-Containing Proteins. Int J Mol Sci 2019; 20:ijms20112760. [PMID: 31195646 PMCID: PMC6600238 DOI: 10.3390/ijms20112760] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
Proteins containing Ly6/uPAR (LU) domains exhibit very diverse biological functions and have broad taxonomic distributions in eukaryotes. In general, they adopt a characteristic three-fingered folding topology with three long loops projecting from a disulfide-rich globular core. The majority of the members of this protein domain family contain only a single LU domain, which can be secreted, glycolipid anchored, or constitute the extracellular ligand binding domain of type-I membrane proteins. Nonetheless, a few proteins contain multiple LU domains, for example, the urokinase receptor uPAR, C4.4A, and Haldisin. In the current review, we will discuss evolutionary aspects of this protein domain family with special emphasis on variations in their consensus disulfide bond patterns. Furthermore, we will present selected cases where missense mutations in LU domain-containing proteins leads to dysfunctional proteins that are causally linked to genesis of human disease.
Collapse
Affiliation(s)
- Julie Maja Leth
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Katrine Zinck Leth-Espensen
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Kristian Kølby Kristensen
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Anni Kumari
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Anne-Marie Lund Winther
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Michael Ploug
- Finsen Laboratory, Ole Maaloes Vej 5, Righospitalet, DK-2200 Copenhagen, Denmark.
- Biotechnology Research Innovation Centre (BRIC), Ole Maaloes Vej 5, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
5
|
Abstract
Glycosylation is a major form of enzymatic modification of organic molecules responsible for multiple biological processes in an organism. The biosynthesis of glycans is controlled by a series of glycosyltransferases, glycosidases and glycan-modifying enzymes that collectively assemble and process monosaccharide moieties into a diverse array of structures. Many studies have provided insight into various pathways of glycosylation at the ocular surface, such as those related to the biosynthesis of mucin-type O-glycans and N-glycans on proteins, but many others still remain largely unknown. This review provides an overview of the different classes of glycans described at the ocular surface focusing on their biosynthetic pathways and biological relevance. A precise understanding of these pathways under physiological and pathological conditions could help identify biomarkers and novel targets for therapeutic intervention.
Collapse
|
6
|
Expression of C4.4A in an In Vitro Human Tissue-Engineered Skin Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2403072. [PMID: 29075641 PMCID: PMC5610857 DOI: 10.1155/2017/2403072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/18/2017] [Indexed: 12/22/2022]
Abstract
A multi-LU-domain-containing protein denoted C4.4A exhibits a tightly regulated membrane-associated expression in the suprabasal layers of stratified squamous epithelia such as skin and the esophagus, and the expression of C4.4A is dysregulated in various pathological conditions. However, the biological function of C4.4A remains unknown. To enable further studies, we evaluated the expression of C4.4A in monolayer cultures of normal human keratinocytes and in tissue-engineered skin substitutes (TESs) produced by the self-assembly approach, which allow the formation of a fully differentiated epidermis tissue. Results showed that, in monolayer, C4.4A was highly expressed in the centre of keratinocyte colonies at cell-cell contacts areas, while some cells located at the periphery presented little C4.4A expression. In TES, emergence of C4.4A expression coincided with the formation of the stratum spinosum. After the creation of a wound within the TES, C4.4A expression was observed in the suprabasal keratinocytes of the migrating epithelium, with the exception of the foremost leading keratinocytes, which were negative for C4.4A. Our results are consistent with previous data in mouse embryogenesis and wound healing. Based on these findings, we conclude that this human TES model provides an excellent surrogate for studies of C4.4A and Haldisin expressions in human stratified epithelia.
Collapse
|
7
|
Görtz M, Galli U, Longerich T, Zöller M, Erb U, Schemmer P. De novo synthesis of C4.4A in hepatocellular carcinoma promotes migration and invasion of tumor cells. Oncol Rep 2017; 38:2697-2704. [PMID: 29048672 PMCID: PMC5780022 DOI: 10.3892/or.2017.5980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 08/31/2017] [Indexed: 01/12/2023] Open
Abstract
C4.4A is a glycoprotein that is upregulated in several human malignancies, including colorectal, breast and renal cell carcinomas. Due to its highly restricted expression in healthy tissue, C4.4A was proposed as a potential diagnostic marker. Thus, the present study was designed to evaluate C4.4A expression and function in hepatocellular carcinoma (HCC) for the first time. Immunohistochemistry was performed to detect expression of C4.4A in human sections of healthy liver, primary HCC in the liver and metastatic HCC in the lung. To assess the contribution of C4.4A to HCC progression proliferation, apoptosis, migration and invasion assays were performed with C4.4A knockdown Huh7 and HepG2 cells. C4.4A is absent in healthy liver tissue. However, intense expression was seen in 59% of primary HCCs and strong expression in 80% of HCC lung metastases. C4.4A expression was also observed in human HCC cell lines, which strongly increased under hypoxic conditions. A C4.4A knock-down revealed that C4.4A is involved in both migration and invasion of HCC cells. Taken together, C4.4A expression in both primary and metastatic HCC suggests its potential value as a diagnostic marker for HCC. Due to its absence in healthy liver tissue, C4.4A might even serve as a possible therapeutic target, particularly for metastatic HCC.
Collapse
Affiliation(s)
- Magdalena Görtz
- Department of General and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Galli
- Department of General and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Margot Zöller
- Tumor Cell Biology, Department of General and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Ulrike Erb
- Tumor Cell Biology, Department of General and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Schemmer
- Department of General and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
8
|
Chen S, Lin L, Yuan C, Gårdsvoll H, Kriegbaum MC, Ploug M, Huang M. Expression and crystallographic studies of the D1D2 domains of C4.4A, a homologous protein to the urokinase receptor. Acta Crystallogr F Struct Biol Commun 2017; 73:486-490. [PMID: 28777093 PMCID: PMC5544007 DOI: 10.1107/s2053230x17009748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/02/2017] [Indexed: 12/12/2022] Open
Abstract
C4.4A is a glycosylphosphatidylinositol-anchored membrane protein comprised of two LU domains (Ly6/uPAR-like domains) and an extensively O-glycosylated C-terminal Ser/Thr/Pro-rich region. C4.4A is a novel biomarker for squamous epithelial differentiation. Its expression is dysregulated under various pathological conditions and it is a robust biomarker for poor prognosis in various malignant conditions such as pulmonary adenocarcinoma. To facilitate crystallization, the two LU domains were excised from intact C4.4A by limited proteolysis, purified and crystallized by the sitting-drop vapour-diffusion method. The crystals diffracted to 2.7 Å resolution and belonged to space group C2221, with unit-cell parameters a = 55.49, b = 119.63, c = 168.54 Å. The statistics indicated good quality of the data, which form a solid basis for the determination of the C4.4A structure.
Collapse
Affiliation(s)
- Shanli Chen
- College of Chemistry, Fuzhou Univerisity, Fuzhou, Fujian 350108, People’s Republic of China
| | - Lin Lin
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Cai Yuan
- College of Bioscience and Biotechnology, Fuzhou Univerisity, Fuzhou, Fujian 350108, People’s Republic of China
| | - Henrik Gårdsvoll
- The Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, Copenhagen University, DK-2200 Copenhagen, Denmark
| | - Mette C. Kriegbaum
- The Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, Copenhagen University, DK-2200 Copenhagen, Denmark
| | - Michael Ploug
- The Finsen Laboratory, Rigshospitalet, DK-2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, Copenhagen University, DK-2200 Copenhagen, Denmark
| | - Mingdong Huang
- College of Chemistry, Fuzhou Univerisity, Fuzhou, Fujian 350108, People’s Republic of China
| |
Collapse
|
9
|
Willuda J, Linden L, Lerchen HG, Kopitz C, Stelte-Ludwig B, Pena C, Lange C, Golfier S, Kneip C, Carrigan PE, Mclean K, Schuhmacher J, von Ahsen O, Müller J, Dittmer F, Beier R, El Sheikh S, Tebbe J, Leder G, Apeler H, Jautelat R, Ziegelbauer K, Kreft B. Preclinical Antitumor Efficacy of BAY 1129980-a Novel Auristatin-Based Anti-C4.4A (LYPD3) Antibody-Drug Conjugate for the Treatment of Non-Small Cell Lung Cancer. Mol Cancer Ther 2017; 16:893-904. [PMID: 28292941 DOI: 10.1158/1535-7163.mct-16-0474] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/11/2016] [Accepted: 02/15/2017] [Indexed: 11/16/2022]
Abstract
C4.4A (LYPD3) has been identified as a cancer- and metastasis-associated internalizing cell surface protein that is expressed in non-small cell lung cancer (NSCLC), with particularly high prevalence in the squamous cell carcinoma (SCC) subtype. With the exception of skin keratinocytes and esophageal endothelial cells, C4.4A expression is scarce in normal tissues, presenting an opportunity to selectively treat cancers with a C4.4A-directed antibody-drug conjugate (ADC). We have generated BAY 1129980 (C4.4A-ADC), an ADC consisting of a fully human C4.4A-targeting mAb conjugated to a novel, highly potent derivative of the microtubule-disrupting cytotoxic drug auristatin via a noncleavable alkyl hydrazide linker. In vitro, C4.4A-ADC demonstrated potent antiproliferative efficacy in cell lines endogenously expressing C4.4A and inhibited proliferation of C4.4A-transfected A549 lung cancer cells showing selectivity compared with a nontargeted control ADC. In vivo, C4.4A-ADC was efficacious in human NSCLC cell line (NCI-H292 and NCI-H322) and patient-derived xenograft (PDX) models (Lu7064, Lu7126, Lu7433, and Lu7466). C4.4A expression level correlated with in vivo efficacy, the most responsive being the models with C4.4A expression in over 50% of the cells. In the NCI-H292 NSCLC model, C4.4A-ADC demonstrated equal or superior efficacy compared to cisplatin, paclitaxel, and vinorelbine. Furthermore, an additive antitumor efficacy in combination with cisplatin was observed. Finally, a repeated dosing with C4.4A-ADC was well tolerated without changing the sensitivity to the treatment. Taken together, C4.4A-ADC is a promising therapeutic candidate for the treatment of NSCLC and other cancers expressing C4.4A. A phase I study (NCT02134197) with the C4.4A-ADC BAY 1129980 is currently ongoing. Mol Cancer Ther; 16(5); 893-904. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kirk Mclean
- Bayer LLC, Mission Bay, San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
LY6/PLAUR domain containing 3 has a role in the maintenance of colorectal cancer stem-like cells. Biochem Biophys Res Commun 2017; 486:232-238. [PMID: 28238780 DOI: 10.1016/j.bbrc.2017.02.112] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/22/2017] [Indexed: 12/15/2022]
Abstract
Colorectal carcinoma (CRC) is one of the most frequently diagnosed cancers and the leading cause of cancer-related death for both men and women. Recent studies have revealed that a small sub-population of cancer cells, termed cancer stem-like cells (CSCs)/cancer-initiating cells (CICs), are endowed with tumor-initiating ability, self-renewal ability and differentiation ability. CSCs/CICs are resistant to current therapies including chemotherapy and radiotherapy. Thus, CSCs/CICs are responsible for recurrence and metastasis, and eradication of CSCs/CICs is essential to cure cancer. In this study, we isolated CR-CSCs/CICs as sphere-cultured cells and found that a product derived from LY6/PLAUR domain containing 3 (LYPD3) is preferentially expressed in CSCs/CICs. Gene overexpression and gene knockdown experiments revealed that LYPD3 has a role in the maintenance of CR-CSCs/CICs. The findings provide a novel molecular insight into CR-CSCs/CICs.
Collapse
|
11
|
Kriegbaum MC, Jacobsen B, Füchtbauer A, Hansen GH, Christensen IJ, Rundsten CF, Persson M, Engelholm LH, Madsen AN, Di Meo I, Lund IK, Holst B, Kjaer A, Lærum OD, Füchtbauer EM, Ploug M. C4.4A gene ablation is compatible with normal epidermal development and causes modest overt phenotypes. Sci Rep 2016; 6:25833. [PMID: 27169360 PMCID: PMC4864438 DOI: 10.1038/srep25833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 04/22/2016] [Indexed: 12/13/2022] Open
Abstract
C4.4A is a modular glycolipid-anchored Ly6/uPAR/α-neurotoxin multidomain protein that exhibits a prominent membrane-associated expression in stratified squamous epithelia. C4.4A is also expressed in various solid cancer lesions, where high expression levels often are correlated to poor prognosis. Circumstantial evidence suggests a role for C4.4A in cell adhesion, migration, and invasion, but a well-defined biological function is currently unknown. In the present study, we have generated and characterized the first C4.4A-deficient mouse line to gain insight into the functional significance of C4.4A in normal physiology and cancer progression. The unchallenged C4.4A-deficient mice were viable, fertile, born in a normal Mendelian distribution and, surprisingly, displayed normal development of squamous epithelia. The C4.4A-deficient mice were, nonetheless, significantly lighter than littermate controls predominantly due to differences in fat mass. Congenital C4.4A deficiency delayed migration of keratinocytes enclosing incisional skin wounds in male mice. In chemically induced bladder carcinomas, C4.4A deficiency attenuated the incidence of invasive lesions despite having no effect on total tumour burden. This new C4.4A-deficient mouse line provides a useful platform for future studies on functional aspects of C4.4A in tumour cell invasion in vivo.
Collapse
Affiliation(s)
- Mette Camilla Kriegbaum
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Jacobsen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Annette Füchtbauer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Gert Helge Hansen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ib Jarle Christensen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Carsten Friis Rundsten
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Morten Persson
- Department of Clinical Physiology, Nuclear Medicine &PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Lars Henning Engelholm
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | - Ivano Di Meo
- Unit of Molecular Neurogenetics, Foundation IRCCS Neurological Institute "Carlo Besta", Milano, Italy
| | - Ida Katrine Lund
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Deparment of Neuroscience and Pharmacology, University of Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine &PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Ole Didrik Lærum
- Department of Pathology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, The Gade Laboratory of Pathology, University of Bergen, Norway
| | | | - Michael Ploug
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Organization, evolution and functions of the human and mouse Ly6/uPAR family genes. Hum Genomics 2016; 10:10. [PMID: 27098205 PMCID: PMC4839075 DOI: 10.1186/s40246-016-0074-2] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/14/2016] [Indexed: 01/08/2023] Open
Abstract
Members of the lymphocyte antigen-6 (Ly6)/urokinase-type plasminogen activator receptor (uPAR) superfamily of proteins are cysteine-rich proteins characterized by a distinct disulfide bridge pattern that creates the three-finger Ly6/uPAR (LU) domain. Although the Ly6/uPAR family proteins share a common structure, their expression patterns and functions vary. To date, 35 human and 61 mouse Ly6/uPAR family members have been identified. Based on their subcellular localization, these proteins are further classified as GPI-anchored on the cell membrane, or secreted. The genes encoding Ly6/uPAR family proteins are conserved across different species and are clustered in syntenic regions on human chromosomes 8, 19, 6 and 11, and mouse Chromosomes 15, 7, 17, and 9, respectively. Here, we review the human and mouse Ly6/uPAR family gene and protein structure and genomic organization, expression, functions, and evolution, and introduce new names for novel family members.
Collapse
|
13
|
Kriegbaum MC, Clausen OPF, Lærum OD, Ploug M. Expression of the Ly6/uPAR-domain proteins C4.4A and Haldisin in non-invasive and invasive skin lesions. J Histochem Cytochem 2014; 63:142-54. [PMID: 25414274 DOI: 10.1369/0022155414563107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
C4.4A and Haldisin belong to the Ly6/uPAR/α-neurotoxin protein domain family. They exhibit highly regulated expression profiles in normal epidermis, where they are confined to early (C4.4A) and late (Haldisin) squamous differentiation. We have now explored if dysregulated expressions occur in non-invasive and invasive skin lesions. In non-invasive lesions, their expression signatures were largely maintained as defined by that of normal epidermis. The scenario was, however, markedly different in the progression towards invasive squamous cell carcinomas. In its non-invasive stage (carcinoma in situ), a pronounced attenuation of C4.4A expression was observed, but upon transition to malignant invasive squamous cell carcinomas, the invasive fronts regained high expression of C4.4A. A similar progression was observed for the early stages of benign infiltrating keratoacanthomas. Interestingly, this transition was accompanied by a shift in the predominant association of C4.4A expression with CK1/10 in the normal epidermis to CK5/14 in the invasive lesions. In contrast, Haldisin expression maintained its confinement to the most-differentiated cells and was hardly expressed in the invasive lesions. Because this altered expression of C4.4A was seen in the invasive front of benign (keratoacanthomas) and malignant (squamous cell carcinomas) neoplasms, we propose that this transition of expression is primarily related to the invasive process.
Collapse
Affiliation(s)
- Mette C Kriegbaum
- The Finsen Laboratory, Rigshospitalet & Biotech Research and Innovation Centre, Copenhagen Biocenter, Copenhagen, Denmark (MCK, MP)
| | - Ole P F Clausen
- TDepartment of Pathology, Oslo University Hospital, University of Oslo, Oslo, Norway (OPFC)
| | - Ole D Lærum
- Department of Pathology, Haukeland University Hospital, Bergen, Norway (ODL)
| | - Michael Ploug
- The Finsen Laboratory, Rigshospitalet & Biotech Research and Innovation Centre, Copenhagen Biocenter, Copenhagen, Denmark (MCK, MP),Danish-Chinese Centre for Proteases and Cancer (MP)
| |
Collapse
|
14
|
Jacobsen B, Kriegbaum MC, Santoni-Rugiu E, Ploug M. C4.4A as a biomarker in pulmonary adenocarcinoma and squamous cell carcinoma. World J Clin Oncol 2014; 5:621-632. [PMID: 25302166 PMCID: PMC4129527 DOI: 10.5306/wjco.v5.i4.621] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 03/10/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023] Open
Abstract
The high prevalence and mortality of lung cancer, together with a poor 5-year survival of only approximately 15%, emphasize the need for prognostic and predictive factors to improve patient treatment. C4.4A, a member of the Ly6/uPAR family of membrane proteins, qualifies as such a potential informative biomarker in non-small cell lung cancer. Under normal physiological conditions, it is primarily expressed in suprabasal layers of stratified squamous epithelia. Consequently, it is absent from healthy bronchial and alveolar tissue, but nevertheless appears at early stages in the progression to invasive carcinomas of the lung, i.e., in bronchial hyperplasia/metaplasia and atypical adenomatous hyperplasia. In the stages leading to pulmonary squamous cell carcinoma, expression is sustained in dysplasia, carcinoma in situ and invasive carcinomas, and this pertains to the normal presence of C4.4A in squamous epithelium. In pulmonary adenocarcinomas, a fraction of cases is positive for C4.4A, which is surprising, given the origin of these carcinomas from mucin-producing and not squamous epithelium. Interestingly, this correlates with a highly compromised patient survival and a predominant solid tumor growth pattern. Circumstantial evidence suggests an inverse relationship between C4.4A and the tumor suppressor LKB1. This might provide a link to the prognostic impact of C4.4A in patients with adenocarcinomas of the lung and could potentially be exploited for predicting the efficacy of treatment targeting components of the LKB1 pathway.
Collapse
|
15
|
Thuma F, Ngora H, Zöller M. The metastasis-associated molecule C4.4A promotes tissue invasion and anchorage independence by associating with the alpha6beta4 integrin. Mol Oncol 2013; 7:917-28. [PMID: 23727360 PMCID: PMC5528461 DOI: 10.1016/j.molonc.2013.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 05/01/2013] [Accepted: 05/02/2013] [Indexed: 01/13/2023] Open
Abstract
C4.4A is a metastasis-associated molecule that functions appear to rely on associated alph6beta4 integrin. To corroborate the impact of the C4.4A-alpha6beta4 integrin association on metastasis formation, C4.4A was knocked-down in a highly metastatic rat pancreatic adenocarcinoma (ASML, ASML-C4.4Akd). Metastasis formation by ASML-C4.4Akd cells after intrafootpad application was strongly retarded in draining nodes and lung colonization was rare. Furthermore, cisplatin treatment significantly prolonged the survival time only of ASML-C4.4Akd-bearing rats. ASML-C4.4Akd cells display reduced migratory activity and impaired matrix protein degradation due to inefficient MMP14 activation; loss of drug-resistance is due to mitigated PI3K/Akt pathway activation. These losses of function rely on the laminin receptor C4.4A recruiting activated alpha6beta4 integrin into rafts, where C4.4A cooperates with alpha6beta4 and via alpha6beta4 with MMP14. Within this raft-located complex, MMP14 provokes focalized matrix degradation and mostly alpha6beta4 integrin promotes BAD phosphorylation and upregulated Bcl2 and BclXl expression. Thus, metastasis-promoting activities of C4.4A are not genuine characteristics of C4.4A. Instead, the raft-located laminin receptor C4.4A recruits alpha6beta4 integrin and supports via the alpha6beta4 integrin MMP14 activation. Thereby C4.4A acts as a linker to facilitate several steps in the metastatic cascade. Taking the restricted C4.4A expression in non-transformed tissue, this knowledge should pave the way toward the use of C4.4A as a therapeutic target.
Collapse
Affiliation(s)
- Florian Thuma
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
| | - Honoré Ngora
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
| | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg D-69120, Germany
- German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
16
|
Gårdsvoll H, Kriegbaum MC, Hertz EP, Alpízar-Alpízar W, Ploug M. The urokinase receptor homolog Haldisin is a novel differentiation marker of stratum granulosum in squamous epithelia. J Histochem Cytochem 2013; 61:802-13. [PMID: 23896969 DOI: 10.1369/0022155413501879] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Several members of the Ly-6/uPAR (LU)-protein domain family are differentially expressed in human squamous epithelia. In some cases, they even play important roles in maintaining skin homeostasis, as exemplified by the secreted single domain member, SLURP-1, the deficiency of which is associated with the development of palmoplantar hyperkeratosis in the congenital skin disorder Mal de Meleda. In the present study, we have characterized a new member of the LU-protein domain family, which we find to be predominantly expressed in the stratum granulosum of human skin, thus resembling the expression of SLURP-1. In accordance with its expression pattern, we denote this protein product, which is encoded by the LYPD5 gene, as Haldisin (human antigen with LU-domains expressed in skin). Two of the five human glycolipid-anchored membrane proteins with multiple LU-domains characterized so far are predominantly confined to squamous epithelia (i.e., C4.4A), to stratum spinosum, and Haldisin to stratum granulosum under normal homeostatic conditions. Whether Haldisin is a prognostic biomarker for certain epithelial malignancies, like C4.4A and SLURP-1, remains to be explored.
Collapse
Affiliation(s)
- Henrik Gårdsvoll
- The Finsen Laboratory, Rigshospitalet & Biotech Research and Innovation Centre, Copenhagen Biocenter, Copenhagen, Denmark (HG,MCK,EPH,WAA,MP)
| | | | | | | | | |
Collapse
|
17
|
Ly6/uPAR-related protein C4.4A as a marker of solid growth pattern and poor prognosis in lung adenocarcinoma. J Thorac Oncol 2013; 8:152-60. [PMID: 23287851 DOI: 10.1097/jto.0b013e318279d503] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION We have recently shown that the protein C4.4A is induced in early precursor lesions of pulmonary adenocarcinomas and squamous cell carcinomas. In the present study, we aimed at analyzing the impact of C4.4A on the survival of non-small cell lung cancer patients and determining whether its unexpected expression in adenocarcinomas could be attributed to a specific growth type (lepidic, acinar, papillary, micropapillary, solid). METHODS Sections from the center and periphery of the primary tumor, as well as N2-positive lymph node metastases, were stained by immunohistochemistry for C4.4A and scored semi-quantitatively for intensity and frequency of positive tumor cells. RESULTS C4.4A score (intensity × frequency) in the tumor center was a highly significant prognostic factor in adenocarcinomas (n = 88), both in univariate (p = 0.004; hazard ratio [95% confidence interval] = 1.44 [1.12-1.85]) and multivariate statistical analysis (p = 0.0005; hazard ratio = 1.65 [1.24-2.19]), demonstrating decreasing survival with increasing score. In contrast, C4.4A did not provide prognostic information in squamous cell carcinomas (n = 104). Pathological stage was significant in both groups. In the adenocarcinomas, C4.4A expression was clearly associated with, but a stronger prognostic factor than, solid growth. CONCLUSIONS The present results substantiate the potential value of C4.4A as a prognostic marker in pulmonary adenocarcinomas seen earlier in a smaller, independent patient cohort. Importantly, we also show that C4.4A is a surrogate marker for adenocarcinoma solid growth. Recent data suggest that C4.4A is negatively regulated by the tumor suppressor liver kinase B1, which is inactivated in some adenocarcinomas, providing a possible link to the impact of C4.4A on the survival of these patients.
Collapse
|