Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 16(10): 2422-2431, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34534447

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) mobilization into the blood occurs under normal physiological conditions and is stimulated in the clinic to enable the isolation of HSPCs for transplantation therapies. In the present study, we identify the tetraspanin CD82 as a novel regulator of HSPC mobilization. Using a global CD82 knockout (CD82KO) mouse, we measure enhanced HSPC mobilization after granulocyte-colony stimulating factor (G-CSF) or AMD3100 treatment, which we find is promoted by increased surface expression of the sphingosine 1-phosphate receptor 1 (S1PR1) on CD82KO HSPCs. Additionally, we identify a disruption in S1PR1 internalization in CD82-deficient HSPCs, suggesting that CD82 plays a critical role in S1PR1 surface regulation. Finally, combining AMD3100 and anti-CD82 treatments, we detect enhanced mobilization of mouse HSPCs and human CD34+ cells in animal models. Together, these data provide evidence that CD82 is an important regulator of HSPC mobilization and suggests exploiting the CD82 scaffold as a therapeutic target to enhance stem cell isolation.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Kangai-1 Protein/physiology , Sphingosine-1-Phosphate Receptors/metabolism , Stem Cells/metabolism , Animals , Antigens, CD34/metabolism , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Tetraspanins/physiology
2.
BMC Cancer ; 20(1): 1211, 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33298014

ABSTRACT

BACKGROUND: Tetraspanin CD82 is a tumor metastasis suppressor that is known to down regulate in various metastatic cancers. However, the exact mechanism by which CD82 prevents cancer metastasis is unclear. This study aims to identify genes that are regulated by CD82 in human prostate cell lines. METHODS: We used whole human genome microarray to obtain gene expression profiles in a normal prostate epithelial cell line that expressed CD82 (PrEC-31) and a metastatic prostate cell line that does not express CD82 (PC3). Then, siRNA silencing was used to knock down CD82 expression in PrEC-31 while CD82 was re-expressed in PC3 to acquire differentially-expressed genes in the respective cell line. RESULTS: Differentially-expressed genes with a P < 0.05 were identified in 3 data sets: PrEC-31 (+CD82) vs PrEC-31(-CD82), PC3-57 (+CD82) vs. PC3-5 V (-CD82), and PC3-29 (+CD82) vs. PC3-5 V (-CD82). Top 25 gene lists did not show overlap within the data sets, except (CALB1) the calcium binding protein calbindin 1 which was significantly up-regulated (2.8 log fold change) in PrEC-31 and PC3-29 cells that expressed CD82. Other most significantly up-regulated genes included serine peptidase inhibitor kazal type 1 (SPINK1) and polypeptide N-acetyl galactosaminyl transferase 14 (GALNT14) and most down-regulated genes included C-X-C motif chemokine ligand 14 (CXCL14), urotensin 2 (UTS2D), and fibroblast growth factor 13 (FGF13). Pathways related with cell proliferation and angiogenesis, migration and invasion, cell death, cell cycle, signal transduction, and metabolism were highly enriched in cells that lack CD82 expression. Expression of two mutually inclusive genes in top 100 gene lists of all data sets, runt-related transcription factor (RUNX3) and trefoil factor 3 (TFF3), could be validated with qRT-PCR. CONCLUSION: Identification of genes and pathways regulated by CD82 in this study may provide additional insights into the role that CD82 plays in prostate tumor progression and metastasis, as well as identify potential targets for therapeutic intervention.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic/genetics , Kangai-1 Protein/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/genetics , Adenocarcinoma/secondary , Cell Line, Tumor , Gene Knockdown Techniques , Gene Ontology , Humans , Kangai-1 Protein/antagonists & inhibitors , Kangai-1 Protein/genetics , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tissue Array Analysis
3.
Geroscience ; 42(2): 595-611, 2020 04.
Article in English | MEDLINE | ID: mdl-32088828

ABSTRACT

Aging is a crucial cause of cognitive decline and a major risk factor for Alzheimer's disease (AD); however, AD's underlying molecular mechanisms remain unclear. Recently, tetraspanins have emerged as important modulators of synaptic function and memory. We demonstrate that the level of tetraspanin CD82 is upregulated in the brains of AD patients and middle-aged mice. In young adult mice, injection of AAV-CD82 to the hippocampus induced AD-like cognitive deficits and impairments in neuronal spine density. CD82 overexpression increased TRPM7 α-kinase cleavage via caspase-3 activation and induced Numb phosphorylation at Thr346 and Ser348 residues. CD82 overexpression promoted beta-amyloid peptide (Aß) secretion which could be reversed by Numb T346S348 mutants. Importantly, hippocampus-related memory functions were improved in Cd82-/- mice. Taken together, our findings provide the evidence that links the elevated CD82-TRPM7-Numb signaling to age-related cognitive impairment.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Kangai-1 Protein , Protein Serine-Threonine Kinases , TRPM Cation Channels , Amyloid beta-Peptides/metabolism , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Humans , Kangai-1 Protein/physiology , Membrane Proteins , Mice , Nerve Tissue Proteins , Protein Serine-Threonine Kinases/physiology , TRPM Cation Channels/physiology
4.
Pediatr Res ; 85(7): 1024-1031, 2019 06.
Article in English | MEDLINE | ID: mdl-30862962

ABSTRACT

BACKGROUND: Stem cell marker CD82 plays a vital role in the oncogenesis and progression of acute myelogenous leukemia (AML), especially in sharing properties of leukemia stem cells (LSCs). The Wnt/ß-catenin pathway is required for the development of LSCs in AML. The present study aimed to validate whether CD82 supports the survival of LSCs in pediatric AML via activation of Wnt/ß-catenin signaling pathway. METHODS: CD82 expression and its correlation with molecules downstream of Wnt/ß-catenin pathway in samples from pediatric AML patients were analyzed. Forced or downregulated expression of CD82 in AML cells was evaluated for the effects of CD82 on cell proliferation, cycle regulation, apoptosis, and adriamycin chemoresistance and to validate the underlying mechanism. RESULT: Aberrant expression of CD82 in pediatric AML patients was found. CD82 messenger RNA expression correlated positively with downstream molecules of Wnt/ß-catenin pathway in AML children. Knockdown of CD82 induced apoptosis, suppressed growth, and decreased adriamycin chemoresistance in AML cells. CD82 accelerated ß-catenin nuclear location and then stimulated the expression of downstream molecules of Wnt/ß-catenin pathway. CONCLUSION: CD82 regulates the proliferation and chemotherapy resistance of AML cells via activation of the Wnt/ß-catenin pathway, which suggest that the CD82 may be a potential therapeutic target in AML children.


Subject(s)
Cell Survival/physiology , Kangai-1 Protein/physiology , Leukemia, Myeloid, Acute/pathology , Wnt Signaling Pathway , beta Catenin/metabolism , Cell Line, Tumor , Child , Humans , Leukemia, Myeloid, Acute/metabolism
5.
Cell Signal ; 52: 83-94, 2018 12.
Article in English | MEDLINE | ID: mdl-30189244

ABSTRACT

Tetraspanin membrane proteins form physical complexes with signaling molecules and have been suggested to influence the signaling events of associated molecules. Of the tetraspanin proteins, CD82 has been shown to promote homotypic cell-cell adhesion, which partially accounts for its role in suppressing cancer invasion and metastasis. We found here that CD82-induced cell-cell adhesion is attributed to increased E-cadherin expression through CD82-mediated downregulation of the E-cadherin repressor Snail. The Snail repression by CD82 resulted from the reduced binding of the Sp1 transcription factor to the Snail gene promoter. Notably, high CD82 expression did not allow the fibronectin matrix to induce Sp1 phosphorylation, implicating CD82 inhibition of the fibronectin-integrin signaling-dependent Sp1 activation. Meanwhile, E-cadherin upregulated by CD82 pulled ß-catenin up to the membrane region, and consequently reduced the amount of cytoplasmic ß-catenin that was able to move into to the nucleus. The Wnt signal-induced nuclear translocation of ß-catenin was also inhibited by the CD82 function of upregulating E-cadherin. Overall, high CD82 expression was likely to suppress fibronectin adhesion-induced Sp1 activation signaling for Snail expression, resulting in continuous E-cadherin expression, which contributed not only to the maintenance of strong cell-cell adhesion but also to the blockage of nuclear ß-catenin signaling.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion , Kangai-1 Protein/physiology , Prostatic Neoplasms/metabolism , Snail Family Transcription Factors/metabolism , Sp1 Transcription Factor/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Fibronectins/metabolism , Humans , Integrins/metabolism , Male , Wnt Signaling Pathway
6.
Ann Rheum Dis ; 77(11): 1619-1626, 2018 11.
Article in English | MEDLINE | ID: mdl-29980577

ABSTRACT

Tetraspanins function as membrane adaptors altering cell-cell fusion, antigen presentation, receptor-mediated signal transduction and cell motility via interaction with membrane proteins including other tetraspanins and adhesion molecules such as integrins. CD82 is expressed in several malignant cells and well described as tumour metastasis suppressor. Rheumatoid arthritis (RA) is based on persistent synovial inflammation and joint destruction driven to a large extent by transformed-appearing activated synovial fibroblasts (SF) with an increased migratory potential. OBJECTIVE: CD82 is upregulated in RA synovial fibroblasts (RASF) compared with osteoarthritis (OA) SF as well as within RA compared with OA synovial lining layer (LL) and the role of CD82 in RASF was evaluated. METHODS: CD82 and integrin immunofluorescence was performed. Lentiviral CD82 overexpression and siRNA-mediated knockdown was confirmed (realtime-PCR, Western blot, immunocytochemistry). RASF migration (Boyden chamber, scrape assay), attachment towards plastic/Matrigel, RASF-binding to endothelial cells (EC) and CD82 expression during long-term invasion in the SCID-mouse-model were evaluated. RESULTS: CD82 was induced by proinflammatory stimuli in SF. In RA-synovium, CD82 was expressed in RASF close to blood vessels, LL, sites of cartilage invasion and colocalised with distinct integrins involved in tumour metastasis suppression but also in RA-synovium by RASF. CD82 overexpression led to reduced RASF migration, cell-matrix and RASF-EC adhesion. Reduced CD82 expression (observed in the sublining) increased RASF migration and matrix adhesion whereas RASF-EC-interaction was reduced. In SCID mice, the presence of CD82 on cartilage-invading RASF was confirmed. CONCLUSION: CD82 could contribute to RASF migration to sites of inflammation and tissue damage, where CD82 keeps aggressive RASF on site.


Subject(s)
Arthritis, Rheumatoid/pathology , Fibroblasts/physiology , Kangai-1 Protein/physiology , Animals , Arthritis, Rheumatoid/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Female , Gene Knockdown Techniques , Humans , Kangai-1 Protein/genetics , Kangai-1 Protein/metabolism , Mice, SCID , RNA, Small Interfering/genetics , Synovial Membrane/metabolism , Synovial Membrane/pathology
7.
J Environ Pathol Toxicol Oncol ; 36(3): 269-275, 2017.
Article in English | MEDLINE | ID: mdl-29283339

ABSTRACT

Lung cancer is the most frequent malignancy and the leading cause of cancer-related death worldwide; it is the second most common cancer, comprising 1.69 million deaths worldwide per year. Among these, 85% of lung cancers are non-small-cell lung carcinoma (NSCLC). Metastasis is common in NSCLC patients and are responsible for most deaths. Kang-Ai 1 (KAI1), a tumor metastasis suppressor gene also known as Cluster of Differentiation 82 (CD82), is a member of the membrane tetraspanin protein family, which are capable of inhibiting the metastatic process in NSCLC. KAI1/CD82 suppresses metastasis via multiple mechanisms regulating inhibition of cell motility, adhesion, fusion, and proliferation. KAI1 may attenuate signaling to shut down metastatic colonization through attenuation of epidermal growth factor receptor (EGFR) signaling and inhibition of the Wnt signaling pathways. In this review, we focus on the differential expression of KAI1/CD82, a tumor metastasis suppressor gene that can inhibit cancer invasion and cell metastasis during NSCLC. The differential expression of KAI1/CD82 could prove to be of novel therapeutic significance in treating malignant tumors and in reducing their metastatic potential.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Genes, Tumor Suppressor , Kangai-1 Protein/genetics , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/physiology , Genes, p53 , Humans , Kangai-1 Protein/antagonists & inhibitors , Kangai-1 Protein/physiology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Wnt Signaling Pathway/physiology
8.
Oncogene ; 36(31): 4381-4392, 2017 08.
Article in English | MEDLINE | ID: mdl-28368419

ABSTRACT

ΔNp63α is a critical mediator of epithelial development and stem cell function in a variety of tissues including the skin and breast, while overexpression of ΔNp63α acts as an oncogene to drive tumor formation and cancer stem cell properties in squamous cell carcinoma. However, with regards to the prostate, while ΔNp63α is expressed in the basal stem cells of the mature gland, during adenocarcinoma development, its expression is lost and its absence is used to clinically diagnose the malignant state. Surprisingly, here we identify a sub-population of bone metastatic prostate cancer cells in the PC3 cell line that express ΔNp63α. Interestingly, we discovered that ΔNp63α favors adhesion and stem-like growth of these cells in the bone microenvironment. In addition, we show that these properties require expression of the target gene CD82. Together, this work uncovers a population of bone metastatic prostate cancer cells that express ΔNp63α, and provides important information about the mechanisms of bone metastatic colonization. Finally, we identify metastasis-promoting properties for the tetraspanin family member CD82.


Subject(s)
Bone Neoplasms/secondary , Kangai-1 Protein/physiology , Prostatic Neoplasms/pathology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Adhesion , Cell Line, Tumor , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL
9.
Oncogene ; 35(31): 4132-40, 2016 08 04.
Article in English | MEDLINE | ID: mdl-26592446

ABSTRACT

Communication between acute myeloid leukemia (AML) and the bone marrow microenvironment is known to control disease progression. Therefore, regulation of AML cell trafficking and adhesion to the bone marrow is of significant interest. In this study, we demonstrate that differential expression of the membrane scaffold CD82 modulates the bone marrow homing of AML cells. By combining mutational analysis and super-resolution imaging, we identify membrane protein clustering by CD82 as a regulator of AML cell adhesion and bone marrow homing. Cluster analysis of super-resolution data indicates that N-linked glycosylation and palmitoylation of CD82 are both critical modifications that control the microdomain organization of CD82 as well as the nanoscale clustering of associated adhesion protein, N-cadherin. We demonstrate that the inhibition of CD82 glycosylation increases the molecular packing of N-cadherin and promotes the bone marrow homing of AML cells. In contrast, we find that the inhibition of CD82 palmitoylation disrupts the formation and organization of N-cadherin clusters and significantly diminishes bone marrow trafficking of AML. Taken together, these data establish a mechanism where the membrane organization of CD82, through specific posttranslational modifications, regulates N-cadherin clustering and membrane density, which impacts the in vivo trafficking of AML cells. As such, these observations provide an alternative model for targeting AML where modulation of protein organization within the membrane may be an effective treatment therapy to disrupt the bone marrow homing potential of AML cells.


Subject(s)
Antigens, CD/chemistry , Bone Marrow/physiology , Cadherins/chemistry , Kangai-1 Protein/physiology , Leukemia, Myeloid, Acute/pathology , Cell Adhesion , Glycosylation , Humans , Lipoylation , Protein Processing, Post-Translational , Receptors, CXCR4/physiology
10.
Leukemia ; 29(12): 2296-306, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26260387

ABSTRACT

The aim of this study was to explore the biological functions of a tetraspanin family protein CD82 expressed aberrantly in chemotherapy-resistant CD34(+)/CD38(-) acute myelogenous leukemia (AML) cells. Microarray analysis of patient-isolated CD34(+)/CD38(-) AML cells revealed that the levels of anti-apoptotic protein BCL2L12 were downregulated after CD82 depletion by specific short hairpin RNA (shRNA). Western blot analysis indicated that BCL2L12 was aberrantly expressed in patient-isolated AML cells and AML cell lines. Furthermore, CD82 blockade by a specific antibody downregulated BCL2L12 in parallel with dephosphorylation of signal transducer and activator of transcription 5 (STAT5) and AKT, whereas pharmacological inhibition of STAT5 and AKT activation decreased BCL2L12 expression in leukemia cells. In addition, shRNA-mediated downregulation of BCL2L12 increased the levels of cleaved caspase-3 and suppressed proliferation of leukemia cells, impairing their engraftment in immunodeficient mice. Taken together, our results indicate that CD82 regulated BCL2L12 expression via STAT5A and AKT signaling and stimulated proliferation and engrafting of leukemia cells, suggesting that CD82 and BCL2L12 may be promising therapeutic targets in AML.


Subject(s)
Kangai-1 Protein/physiology , Leukemia, Myeloid, Acute/pathology , Muscle Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , STAT5 Transcription Factor/physiology , Signal Transduction/physiology , Aged , Aged, 80 and over , Apoptosis , Female , Humans , Kangai-1 Protein/analysis , Male , Middle Aged , Muscle Proteins/analysis , Proto-Oncogene Proteins c-bcl-2/analysis , Transcriptome
11.
Mol Carcinog ; 53(8): 610-24, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23401136

ABSTRACT

The KAI1/CD82 tetraspanin is a widely expressed cell surface molecule thought to organize diverse cellular signaling processes. KAI1/CD82 suppresses metastasis but not tumorigenicity, establishing it as one of a class of metastasis suppressor genes. In order to further assess its functions, we have characterized the phenotypic properties of Kai1/Cd82 deleted mice, including viability, fertility, lymphocyte composition, blood chemistry and tissue histopathology, and of their wild-type and heterozygote littermates. Interestingly, Kai1/Cd82(-/-) showed no obvious genotype associated defects in any of these processes and displayed no genotype associated histopathologic abnormalities after 12 or 18 months of life. Expression profiles of non-immortal, wild-type and Kai1/Cd82(-/-) mouse embryo fibroblast (MEFs) indicated distinct sex-specific and genotype-specific profiles. These data identify 191 and 1,271 differentially expressed transcripts (by twofold at P < 0.01) based on Kai1/CD82 genotype status in female and male MEFs, respectively. Differentially expressed genes in male MEFs were surprisingly enriched for cell division related processes, suggesting that Kai1/Cd82 may functionally affect these processes. This suggests that Kai/Cd82 has an unappreciated role in the early establishment of proliferation and division when challenged with a new environment that might play a role in adaptability to new metastatic sites.


Subject(s)
Cell Proliferation , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Kangai-1 Protein/physiology , Neoplasms, Experimental/mortality , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Northern , Blotting, Western , Cells, Cultured , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
12.
FEBS Lett ; 586(22): 4031-7, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23068610

ABSTRACT

The metastasis suppressor KAI1/CD82 has been implicated in various cellular processes; however, its function in development is not fully understood. Here, we generated and characterized mutants of Tsp66E and Tsp74F, which are Drosophila homologues of KAI1/CD82 and Tspan11, respectively. These mutants exhibited egg elongation defects along with disturbed integrin localization and actin polarity. Moreover, the defects were enhanced by mutation of inflated, an αPS2 integrin gene. Mutant ovaries had elevated αPS2 integrin levels and reduced endocytic trafficking. These results suggest that Drosophila KAI1/CD82 affects the polarized localization and the level of integrin, which may contribute to epithelial cell polarity.


Subject(s)
Drosophila Proteins/physiology , Integrin alpha Chains/physiology , Integrins/metabolism , Ovarian Follicle/growth & development , Tetraspanins/physiology , Wings, Animal/growth & development , Actins/metabolism , Animals , Blotting, Western , Dextrans/metabolism , Dextrans/pharmacokinetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Endocytosis , Female , Humans , Immunohistochemistry , Integrin alpha Chains/genetics , Kangai-1 Protein/genetics , Kangai-1 Protein/physiology , Male , Microscopy, Confocal , Mutation , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovum/growth & development , Ovum/metabolism , Tetraspanins/genetics , Wings, Animal/metabolism
13.
BMC Cancer ; 12: 81, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22390300

ABSTRACT

BACKGROUND: KAI1 was initially identified as a metastasis-suppressor gene in prostate cancer. It is a member of the tetraspan transmembrane superfamily (TM4SF) of membrane glycoproteins. As part of a tetraspanin-enriched microdomain (TEM), KAI1 inhibits tumor metastasis by negative regulation of Src. However, the underlying regulatory mechanism has not yet been fully elucidated. CUB-domain-containing protein 1 (CDCP1), which was previously known as tetraspanin-interacting protein in TEM, promoted metastasis via enhancement of Src activity. To better understand how KAI1 is involved in the negative regulation of Src, we here examined the function of KAI1 in CDCP1-mediated Src kinase activation and the consequences of this process, focusing on HIF-1 α and VEGF expression. METHODS: We used the human prostate cancer cell line PC3 which was devoid of KAI1 expression. Vector-transfected cells (PC3-GFP clone #8) and KAI1-expressing PC3 clones (PC3-KAI1 clone #5 and #6) were picked after stable transfection with KAI1 cDNA and selection in 800 µg/ml G418. Protein levels were assessed by immunoblotting and VEGF reporter gene activity was measured by assaying luciferase activitiy. We followed tumor growth in vivo and immunohistochemistry was performed for detection of HIF-1, CDCP1, and VHL protein level. RESULTS: We demonstrated that Hypoxia-inducible factor 1α (HIF-1α) and VEGF expression were significantly inhibited by restoration of KAI1 in PC3 cells. In response to KAI1 expression, CDCP1-enhanced Src activation was down-regulated and the level of von Hippel-Lindau (VHL) protein was significantly increased. In an in vivo xenograft model, KAI1 inhibited the expression of CDCP1 and HIF-1α. CONCLUSIONS: These novel observations may indicate that KAI1 exerts profound metastasis-suppressor activity in the tumor malignancy process via inhibition of CDCP1-mediated Src activation, followed by VHL-induced HIF-1α degradation and, ultimately, decreased VEGF expression.


Subject(s)
Antigens, CD/physiology , Cell Adhesion Molecules/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kangai-1 Protein/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/metabolism , Antigens, Neoplasm , Cell Line, Tumor , Cell Movement , Humans , Immunoblotting , Immunohistochemistry , Kangai-1 Protein/metabolism , Luciferases/metabolism , Male , Neoplasm Invasiveness , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
14.
Biochem Biophys Res Commun ; 415(4): 619-26, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22079629

ABSTRACT

Tetraspanins have gained increased attention due to their functional versatility. But the universal cellular mechanism that governs such versatility remains unknown. Herein we present the evidence that tetraspanins CD81 and CD82 regulate the formation and/or development of cell membrane protrusions. We analyzed the ultrastructure of the cells in which a tetraspanin is either overexpressed or ablated using transmission electron microscopy. The numbers of microvilli on the cell surface were counted, and the radii of microvillar tips and the lengths of microvilli were measured. We found that tetraspanin CD81 promotes the microvillus formation and/or extension while tetraspanin CD82 inhibits these events. In addition, CD81 enhances the outward bending of the plasma membrane while CD82 inhibits it. We also found that CD81 and CD82 proteins are localized at microvilli using immunofluorescence. CD82 regulates microvillus morphogenesis likely by altering the plasma membrane curvature and/or the cortical actin cytoskeletal organization. We predict that membrane protrusions embody a common morphological phenotype and cellular mechanism for, at least some if not all, tetraspanins. The differential effects of tetraspanins on microvilli likely lead to the functional diversification of tetraspanins and appear to correlate with their functional propensity.


Subject(s)
Cell Membrane/physiology , Kangai-1 Protein/physiology , Tetraspanin 28/physiology , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Kangai-1 Protein/genetics , Kangai-1 Protein/metabolism , Mice , Mice, Mutant Strains , Microvilli/physiology , Microvilli/ultrastructure , Tetraspanin 28/genetics , Tetraspanin 28/metabolism
15.
Hepatobiliary Pancreat Dis Int ; 10(2): 201-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21459729

ABSTRACT

BACKGROUND: KAI1/CD82 has been reported to attenuate the process of metastases in a variety of tumors; however, its mechanism of action in invasion has not been fully elucidated. The present study aimed to investigate the importance of KAI1 in invasion and its correlation with activation of sphingosine kinase (SPK) in human pancreatic cancer PANC1 and Miapaca-2 cell lines. METHODS: The expression of KAI1 in PANC1 and Miapaca-2 cells, which was mediated by recombinant adenovirus (Ad-KAI1), was assessed by a flow cytometer and Western blotting. After successful infection was established, in vitro growth curve and invasive ability in Boyden Chamber assay were studied. The presence of KAI1 correlating with c-Met and SPK was detected by co-immunoprecipitation and [gamma-32P] ATP incorporation. RESULTS: KAI1 genes had no significant effects on the curve representing cell growth. After infection with the KAI1 gene, decreased invasive ability in the Boyden Chamber assay was observed in PANC1 and Miapaca-2 cells that were induced by hepatocyte growth factor. Over-expression of KAI1 in the cells led to the deactivation of SPK and a decreased level of intracellular sphingosine-1-phosphate. No correlation was observed between c-Met and KAI1 during co-immunoprecipitation. CONCLUSION: The results of this study for the first time demonstrated a regulatory role for KAI1 in SPK activation, which leads to decreased invasive ability in disease progression of human pancreatic cancer.


Subject(s)
Hepatocyte Growth Factor/antagonists & inhibitors , Kangai-1 Protein/physiology , Pancreatic Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adenoviridae/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Hepatocyte Growth Factor/physiology , Humans , Kangai-1 Protein/genetics , Neoplasm Invasiveness , Proto-Oncogene Proteins c-met/physiology , Transfection
16.
J Neurosci ; 29(36): 11172-81, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19741124

ABSTRACT

In the adult mammalian brain, oligodendrocyte progenitors can differentiate into mature oligodendrocytes during remyelination. Mechanisms that regulate migration and differentiation of progenitors are of great importance in understanding normal development and demyelinating/remyelinating conditions. In a microarray analysis comparing adult and neonatal O4-positive (+) cells, we found that the tetraspanin KAI1/CD82 is far more highly expressed in adult O4(+) cells than in neonatal O4(+) cells (Lin et al., 2009). CD82 is a metastasis suppressor, and its expression is often downregulated or lost in the advanced stages of metastatic cancer. We hypothesized that CD82 could be a factor that restricts migration and promotes differentiation of maturing oligodendrocytes. Western blot analysis of isolated adult O4(+) cells confirms the elevated levels of CD82, which continues to be expressed as these become O1(+) in vitro. In the adult rat white matter, CD82 is coexpressed with CC1 and olig2 but not with NG2 or GFAP. Immature cells of the neonatal forebrain subventricular zone (SVZ) infected in vivo with a retrovirus that constitutively expresses CD82 do not remain immature but differentiate into either CC1(+) and MBP(+) myelinating oligodendrocytes in the white matter or zebrinII(+) astrocytes in the cortex. Their migration from the SVZ is severely restricted. In contrast, downregulation of CD82 in SVZ cells in vivo, using retroviral-expressed short hairpin RNAs (shRNAs), prevents their differentiation into myelinating oligodendrocytes. shRNA-expressing cells remained PDGF receptor alpha positive, olig2(+), or NG2(+) or became CC1(+) nonmyelinating oligodendrocytes or GFAP(+) astrocytes. CD82 thus appears to be a critical molecule in the regulation of oligodendrocyte progenitor migration and myelination.


Subject(s)
Cell Differentiation/genetics , Cell Movement/genetics , Kangai-1 Protein/biosynthesis , Myelin Sheath/genetics , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Cell Lineage/genetics , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Kangai-1 Protein/genetics , Kangai-1 Protein/physiology , Myelin Sheath/physiology , Neurogenesis/genetics , Oligodendroglia/cytology , Rats , Rats, Sprague-Dawley , Stem Cells/cytology
17.
Br J Cancer ; 101(8): 1402-9, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19755988

ABSTRACT

BACKGROUND: Multiple myeloma (MM) therapy is hindered by the interaction of the heterogeneous malignant plasma cells with their microenvironment and evolving drug resistance. We have previously shown that the membranal tetraspanins, CD81 and CD82, are under-expressed in MM cells and that their reintroduction causes massive non-apoptotic death. In this study, we aimed to characterise the tetraspanin-induced MM death. METHODS: Multiple myeloma cell lines were transiently transfected with eGFP-CD81N1/CD82N1 fusion proteins and assessed for death mode by flow cytometry (propidium iodide, ZVAD-fmk, 3MA), activation of unfolded protein response (UPR), and autophagy (immunoblot, RT-PCR). RESULTS: Cell death induced by CD81N1 and CD82N1 in MM cell lines was autophagic and involved endoplasmic reticulum (ER)-stress manifested by activation of UPR pathways, PERK (protein kinase-like ER kinase) and IRE1 (inositol-requiring 1). We also established the relative X-box binding protein 1 baseline expression levels in a panel of MM cell lines and their general dependence on autophagy for survival. Timeline of UPR cascades and cell fate supported our results. INTERPRETATION: This is the first publication implicating tetraspanins in UPR signalling pathways, autophagy, and autophagic death. Integration of our findings with published data highlights the unifying dependence of MM cells on ER-Golgi homoeostasis, and underscores the potential of tetraspanin complexes and ER-stress as leverage for MM therapy.


Subject(s)
Antigens, CD/physiology , Apoptosis , Autophagy , Endoplasmic Reticulum/metabolism , Kangai-1 Protein/physiology , Multiple Myeloma/pathology , Protein Folding , Signal Transduction , Cell Line, Tumor , Cell Survival , Humans , Multiple Myeloma/therapy , Tetraspanin 28
18.
Histol Histopathol ; 24(4): 519-30, 2009 04.
Article in English | MEDLINE | ID: mdl-19224455

ABSTRACT

CD82, also known as KAI-1, structurally belongs to tetraspanin family while categorised as metastasis suppressor gene on functional grounds. KAI1/CD82 is localized on cell membrane and form interactions with other tetraspanins, integrins and chemokines which are respectively responsible for cell migration, adhesion and signalling. In recent years apart from its significant involvement in the suppression of secondary tumours it has also been observed that KAI1/CD82 plays a vital role in virus binding and its entry inside the cell. Decreased expression of KAI1/CD82 molecule results in aggravating cancer progression. Altered expression levels of KAI1/CD82 molecule in different types of human cancer have been implicated as having prognostic value and linking to the long term survival of the patients. Increased level of KAI1/CD82 also results in the suppression of secondary tumour growth. Increased expression of this molecule results in reduced cell invasion and cell migration due to endocytosis of epidermal growth factor receptors (EGFR). Thus, KAI-1/CD82 is a pivotal molecule in the regulation of cancer cells' behaviour and has important clinical and therapeutic implications in cancer.


Subject(s)
Kangai-1 Protein/physiology , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology , Biomarkers, Tumor , Cell Transformation, Neoplastic , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy , Pregnancy , Signal Transduction , Virus Attachment
19.
Nat Rev Cancer ; 9(1): 40-55, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19078974

ABSTRACT

Tumours progress through a cascade of events that enable the formation of metastases. Some of the components that are required for this fatal process are well established. Tetraspanins, however, have only recently received attention as both metastasis suppressors and metastasis promoters. This late appreciation is probably due to their capacity to associate with various molecules, which they recruit into special membrane microdomains, and their abundant presence in tumour-derived small vesicles that aid intercellular communication. It is reasonable to assume that differences in the membrane and vesicular web components that associate with individual tetraspanins account for their differing abilities to promote and suppress metastasis.


Subject(s)
Membrane Proteins/physiology , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/physiology , Animals , Antigens, CD/physiology , Antigens, Neoplasm/physiology , Cell Communication , Cell Movement/physiology , Disease Progression , Exosomes/physiology , Humans , Integrins/physiology , Kangai-1 Protein/physiology , Membrane Glycoproteins/physiology , Membrane Microdomains/physiology , Membrane Microdomains/ultrastructure , Membrane Proteins/chemistry , Membrane Proteins/genetics , Multigene Family , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neovascularization, Pathologic/physiopathology , Protein Structure, Tertiary , Rats , Structure-Activity Relationship , Tetraspanin 24 , Tetraspanin 29 , Tetraspanins
20.
Cancer Lett ; 266(2): 163-70, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18395972

ABSTRACT

In this study, we analyzed the effect of the metastasis suppressor CD82/KAI-1, a member of the tetraspanin superfamily, on intercellular adhesion on cancer cells. The newly established invasion assay and the cell aggregation assay revealed that CD82 strengthens E-cadherin-mediated intercellular adhesion. Interestingly, ectopic expression of CD82 stabilized E-cadherin/beta-catenin complex formation. Furthermore, CD82 reduced tyrosine phosphorylation of beta-catenin on HGF stimulation. Taken together, CD82 may stabilize or strengthen E-cadherin-dependent intercellular adhesion by regulating beta-catenin-mediated signal transduction on cancer cells, and consequently, prevent cancer cells from seceding from the primary tumor site.


Subject(s)
Cadherins/metabolism , Kangai-1 Protein/physiology , Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Humans , Neoplasms/pathology , Phosphorylation , Tyrosine/metabolism , beta Catenin/chemistry , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...