Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 933
Filter
1.
Int J Mol Sci ; 24(4)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36835416

ABSTRACT

CD44 is a cell surface glycoprotein, and its isoforms are produced by the alternative splicing with the standard and variant exons. The CD44 variant exon-containing isoforms (CD44v) are overexpressed in carcinomas. CD44v6 is one of the CD44v, and its overexpression predicts poor prognosis in colorectal cancer (CRC) patients. CD44v6 plays critical roles in CRC adhesion, proliferation, stemness, invasiveness, and chemoresistance. Therefore, CD44v6 is a promising target for cancer diagnosis and therapy for CRC. In this study, we established anti-CD44 monoclonal antibodies (mAbs) by immunizing mice with CD44v3-10-overexpressed Chinese hamster ovary (CHO)-K1 cells. We then characterized them using enzyme-linked immunosorbent assay, flow cytometry, western blotting, and immunohistochemistry. One of the established clones (C44Mab-9; IgG1, kappa) reacted with a peptide of the variant 6-encoded region, indicating that C44Mab-9 recognizes CD44v6. Furthermore, C44Mab-9 reacted with CHO/CD44v3-10 cells or CRC cell lines (COLO201 and COLO205) by flow cytometry. The apparent dissociation constant (KD) of C44Mab-9 for CHO/CD44v3-10, COLO201, and COLO205 was 8.1 × 10-9 M, 1.7 × 10-8 M, and 2.3 × 10-8 M, respectively. C44Mab-9 detected the CD44v3-10 in western blotting, and partially stained the formalin-fixed paraffin-embedded CRC tissues in immunohistochemistry. Collectively, C44Mab-9 is useful for detecting CD44v6 in various applications.


Subject(s)
Antibodies, Monoclonal , Colorectal Neoplasms , Hyaluronan Receptors , Animals , Cricetinae , Mice , CHO Cells , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/therapy , Cricetulus , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Receptors/immunology , Protein Isoforms/metabolism
2.
Lab Invest ; 102(2): 172-184, 2022 02.
Article in English | MEDLINE | ID: mdl-34782726

ABSTRACT

The phenotype of glioma-initiating cells (GIC) is modulated by cell-intrinsic and cell-extrinsic factors. Phenotypic heterogeneity and plasticity of GIC is an important limitation to therapeutic approaches targeting cancer stem cells. Plasticity also presents a challenge to the identification, isolation, and propagation of purified cancer stem cells. Here we use a barcode labelling approach of GIC to generate clonal populations over a number of passages, in combination with phenotyping using the established stem cell markers CD133, CD15, CD44, and A2B5. Using two cell lines derived from isocitrate dehydrogenase (IDH)-wildtype glioblastoma, we identify a remarkable heterogeneity of the phenotypes between the cell lines. During passaging, clonal expansion manifests as the emergence of a limited number of barcoded clones and a decrease in the overall number of clones. Dual-labelled GIC are capable of forming traceable clonal populations which emerge after as few as two passages from mixed cultures and through analyses of similarity of relative proportions of 16 surface markers we were able to pinpoint the fate of such populations. By generating tumour organoids we observed a remarkable persistence of dominant clones but also a significant plasticity of stemness marker expression. Our study presents an experimental approach to simultaneously barcode and phenotype glioma-initiating cells to assess their functional properties, for example to screen newly established GIC for tumour-specific therapeutic vulnerabilities.


Subject(s)
Antigens, CD/immunology , Brain Neoplasms/immunology , Glioma/immunology , Neoplastic Stem Cells/immunology , Tumor Microenvironment/immunology , AC133 Antigen/immunology , AC133 Antigen/metabolism , Antigens, CD/metabolism , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Clone Cells/immunology , Clone Cells/metabolism , Flow Cytometry , Glioma/metabolism , Glioma/pathology , Humans , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Immunophenotyping , Lewis X Antigen/immunology , Lewis X Antigen/metabolism , Microscopy, Confocal , Neoplastic Stem Cells/classification , Neoplastic Stem Cells/metabolism
3.
Front Immunol ; 12: 782448, 2021.
Article in English | MEDLINE | ID: mdl-34868059

ABSTRACT

Background: Treatment of B-cell malignancies with CD19-directed chimeric antigen receptor (CAR) T-cells marked a new era in immunotherapy, which yet has to be successfully adopted to solid cancers. Epigenetic inhibitors of DNA methyltransferases (DNMTi) and histone deacetylases (HDACi) can induce broad changes in gene expression of malignant cells, thus making these inhibitors interesting combination partners for immunotherapeutic approaches. Methods: Urothelial carcinoma cell lines (UCC) and benign uroepithelial HBLAK cells pretreated with the DNMTi decitabine or the HDACi romidepsin were co-incubated with CAR T-cells directed against EGFR or CD44v6, and subsequent cytotoxicity assays were performed. Effects on T-cell cytotoxicity and surface antigen expression on UCC were determined by flow cytometry. We also performed next-generation mRNA sequencing of inhibitor-treated UCC and siRNA-mediated knockdown of potential regulators of CAR T-cell killing. Results: Exposure to decitabine but not romidepsin enhanced CAR T-cell cytotoxicity towards all UCC lines, but not towards the benign HBLAK cells. Increased killing could neither be attributed to enhanced target antigen expression (EGFR and CD44v6) nor fully explained by changes in the T-cell ligands PD-L1, PD-L2, ICAM-1, or CD95. Instead, gene expression analysis suggested that regulators of cell survival and apoptosis were differentially induced by the treatment. Decitabine altered the balance between survival and apoptosis factors towards an apoptosis-sensitive state associated with increased CAR T-cell killing, while romidepsin, at least partially, tilted this balance in the opposite direction. Knockdown experiments with siRNA in UCC confirmed BID and BCL2L1/BCLX as two key factors for the altered susceptibility of the UCC. Conclusion: Our data suggest that the combination of decitabine with CAR T-cell therapy is an attractive novel therapeutic approach to enhance tumor-specific killing of bladder cancer. Since BID and BCL2L1 are essential determinants for the susceptibility of a wide variety of malignant cells, their targeting might be additionally suitable for combination with immunotherapies, e.g., CAR T-cells or checkpoint inhibitors in other malignancies.


Subject(s)
Epigenesis, Genetic , Hyaluronan Receptors/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Apoptosis , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Hyaluronan Receptors/immunology , Immunomodulation , Immunophenotyping , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction , Treatment Outcome , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/therapy
4.
Virulence ; 12(1): 2352-2365, 2021 12.
Article in English | MEDLINE | ID: mdl-34515624

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is emerging as a major threat to the global swine industry. Clinical PEDV infection is associated with severe intestinal lesions, resulting in absorptive dysfunction and high mortality rates in suckling piglets. The extracellular matrix (ECM) is an important component of intestinal tissue, providing a structural framework and conveying tissue-specific signals to nearby enterocytes. In this study, we investigated the extensive ECM remodeling observed in intestinal epithelial cells infected with PEDV and elucidated the associated activated ECM receptor-related pathways. Protein-protein interaction network analysis revealed two significantly differentially expressed genes (cluster of differentiation 44 [CD44] and serpin family E member 1 [SERPINE1]) associated with the ECM. At the transcriptional level, both genes exhibited significant positive correlation with the extent of PEDV replication. Similarly, the expression of CD44 and PAI-1 (encoded by SERPINE1) was also increased in the intestines of piglets during viral infection. Furthermore, CD44 exhibited antiviral activity by enhancing the expression of antiviral cytokines (e.g., interleukin [IL]-6, IL-18, IL-11, and antimicrobial peptide beta-defensin 1) by activating nuclear factor-κB signaling. Conversely, PAI-1 was found to promote the release of progeny virions during PEDV infection, despite a decreased intracellular viral load. Nevertheless, the underlying mechanisms are still unclear. Taken together, our results highlighted the biological roles of specific ECM-regulated genes, i.e., CD44 and SERPINE1 in suppressing and promoting PEDV infection, thereby providing a theoretical foundation for the role of the ECM in intestinal infections and identifying potential therapeutic targets for PEDV.


Subject(s)
Coronavirus Infections , Extracellular Matrix , Signal Transduction , Swine Diseases , Animals , Antimicrobial Peptides/immunology , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Hyaluronan Receptors/immunology , Intestines/virology , Plasminogen Activator Inhibitor 1/immunology , Porcine epidemic diarrhea virus , Swine , Swine Diseases/immunology , Swine Diseases/virology
5.
Int J Mol Sci ; 22(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34502547

ABSTRACT

Cancer stem cells (CSCs) can be induced from differentiated cancer cells in the tumor microenvironment or in response to treatments and exhibit chemo- and radioresistance, leading to tumor recurrence and metastasis. We previously reported that triple negative breast cancer (TNBC) cells with acquired radioresistance exhibited more aggressive features due to an increased CSC population. Therefore, here, we isolated CSCs from radiotherapy-resistant (RT-R)-TNBC cells and investigated the effects of these CSCs on tumor progression and NK cell-mediated cytotoxicity. Compared to MDA-MB-231 and RT-R-MDA-MB-231 cells, CD24-/low/CD44+ cells isolated from RT-R-MDA-MB-231 cells showed increased proliferation, migration and invasion abilities, and induced expression of tumor progression-related molecules. Moreover, similar to MDA-MB-231 cells, CD24-/low/CD44+ cells recruited NK cells but suppressed NK cell cytotoxicity by regulating ligands for NK cell activation. In an in vivo model, CD24-/low/CD44+ cell-injected mice showed enhanced tumor progression and lung metastasis via upregulation of tumor progression-related molecules and altered host immune responses. Specifically, NK cells were recruited into the peritumoral area tumor but lost their cytotoxicity due to the altered expression of activating and inhibitory ligands on tumors. These results suggest that CSCs may cause tumor evasion of immune cells, resulting in tumor progression.


Subject(s)
Breast Neoplasms/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Neoplastic Stem Cells/immunology , Xenograft Model Antitumor Assays/methods , Animals , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , CD24 Antigen/immunology , CD24 Antigen/metabolism , Cell Line , Cell Line, Tumor , Cell Movement/immunology , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/radiation effects , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Radiotherapy/methods
6.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34088845

ABSTRACT

CD8+ T cells play a central role in antitumor immune responses that kill cancer cells directly. In aged individuals, CD8+ T cell immunity is strongly suppressed, which is associated with cancer and other age-related diseases. The mechanism underlying this age-related decrease in immune function remains largely unknown. This study investigated the role of T cell function in age-related unresponsiveness to PD-1 blockade cancer therapy. We found inefficient generation of CD44lowCD62Llow CD8+ T cell subset (P4) in draining lymph nodes of tumor-bearing aged mice. In vitro stimulation of naive CD8+ T cells first generated P4 cells, followed by effector/memory T cells. The P4 cells contained a unique set of genes related to enzymes involved in one-carbon (1C) metabolism, which is critical to antigen-specific T cell activation and mitochondrial function. Consistent with this finding, 1C-metabolism-related gene expression and mitochondrial respiration were down-regulated in aged CD8+ T cells compared with young CD8+ T cells. In aged OVA-specific T cell receptor (TCR) transgenic mice, ZAP-70 was not activated, even after inoculation with OVA-expressing tumor cells. The attenuation of TCR signaling appeared to be due to elevated expression of CD45RB phosphatase in aged CD8+ T cells. Surprisingly, strong stimulation by nonself cell injection into aged PD-1-deficient mice restored normal levels of CD45RB and ameliorated the emergence of P4 cells and 1C metabolic enzyme expression in CD8+ T cells, and antitumor activity. These findings indicate that impaired induction of the P4 subset may be responsible for the age-related resistance to PD-1 blockade, which can be rescued by strong TCR stimulation.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Hyaluronan Receptors/immunology , L-Selectin/immunology , Neoplasms, Experimental/immunology , Aging/genetics , Animals , Cell Line, Tumor , Hyaluronan Receptors/genetics , L-Selectin/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/immunology
7.
J Cell Mol Med ; 25(14): 6634-6651, 2021 07.
Article in English | MEDLINE | ID: mdl-34075722

ABSTRACT

Available therapies aimed at treating age-related osteoporosis are still insufficient. Therefore, designing reliable in vitro model for the analysis of molecular mechanisms underlying senile osteoporosis is highly required. We have isolated and characterized progenitor cells isolated from bone marrow (BMSCs) of osteoporotic mice strain SAM/P6 (BMSCSAM/P6 ). The cytophysiology of BMSCSAM/P6 was for the first time compared with BMSCs isolated from healthy BALB/c mice (BMSCBALB/c ). Characterization of the cells included evaluation of their multipotency, morphology and determination of specific phenotype. Viability of BMSCs cultures was determined in reference to apoptosis profile, metabolic activity, oxidative stress, mitochondrial membrane potential and caspase activation. Additionally, expression of relevant biomarkers was determined with RT-qPCR. Obtained results indicated that BMSCSAM/P6 and BMSCBALB/c show the typical phenotype of mesenchymal stromal cells (CD44+, CD73+, CD90+) and do not express CD45. Further, BMSCSAM/P6 were characterized by deteriorated multipotency, decreased metabolic activity and increased apoptosis occurrence, accompanied by elevated oxidative stress and mitochondria depolarisation. The transcriptome analyses showed that BMSCSAM/P6 are distinguished by lowered expression of molecules crucial for proper osteogenesis, including Coll-1, Opg and Opn. However, the expression of Trap, DANCR1 and miR-124-3p was significantly up-regulated. Obtained results show that BMSCSAM/P6 present features of progenitor cells with disturbed metabolism and could serve as appropriate model for in vitro investigation of age-dependent osteoporosis.


Subject(s)
Cell Differentiation/genetics , Mesenchymal Stem Cells/immunology , Osteogenesis/genetics , Osteoporosis/genetics , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , Animals , Cell Differentiation/immunology , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Osteoblasts/immunology , Osteoblasts/metabolism , Osteogenesis/immunology , Osteoporosis/immunology , Osteoporosis/pathology , Stem Cells/immunology , Stem Cells/metabolism , Thy-1 Antigens/genetics , Thy-1 Antigens/immunology
8.
Sci Rep ; 11(1): 9708, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33958632

ABSTRACT

In this study, we set out to identify regulators of intact amyloid-ß40/42 (Aß) levels in A549 (p53 wild-type) and H1299 (p53-null) lung cancer cell media. Higher Aß levels were detected in the media of A549 than H1299 cells without or with treatment with 4-methylumbelliferone (4-MU) and/or the anti-CD44 antibody (5F12). Using inhibitors, we found that PI3K, AKT, and NFκB are likely involved in regulating Aß levels in the media. However, increased Aß levels that more closely resembled those found upon 4-MU co-treatment resulted from MMP2/9 inhibition, suggesting that MMP2/9 maybe the main contributors to regulation of Aß levels in the media. Differences in Aß levels might be accounted for, in part, by p53 since blocking p53 function in A549 cells resulted in decreased Aß levels, increased MMP2/9 levels, increased PI3K/AKT activities and the phospho/total NFκB ratio. Using siRNA targeted against MMP2 or MMP9, we found increased Aß levels in the media, however, MMP2 knockdown led to Aß levels closely mimicking those detected by co-treatment with 4-MU. Cell viability or apoptosis upon treatment with either MMP2 or MMP9 siRNA along with Aß immunodepletion, showed that MMP2 is the predominant regulator of the cytotoxic effects induced by Aß in lung cancer cells.


Subject(s)
Amyloid beta-Peptides/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Benzothiazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Culture Media, Conditioned , Humans , Hyaluronan Receptors/immunology , Hymecromone/pharmacology , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/genetics , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Toluene/analogs & derivatives , Toluene/pharmacology
9.
Oral Oncol ; 116: 105259, 2021 05.
Article in English | MEDLINE | ID: mdl-33895463

ABSTRACT

Immune checkpoint blockade can cause regression of recurrent and/or refractory head and neck squamous cell carcinoma (HNSCC). As a second type of immunotherapy, adoptive cellular therapy with genetically modified patient's T-cells redirected against the autologous malignant cells by expressing chimeric antigen receptors (CARs) recognizing tumor-associated antigens has been established as highly efficient personalized treatment for hematological malignancies. In solid cancers however, the application of these genetically modified immune effector cells still lacks equal response rates. CD44v6 is an isoform of the hyaluronic receptor CD44 that is almost exclusively expressed at high levels on solid cancers and has been associated with tumorigenesis, tumor cell invasion and metastasis. Here, we established a highly specific CAR against CD44v6 on HNSCC cells that can be expressed on normal T-cells with lentiviral vectors. Using primary human HNSCC cells in combination with CRISPR/Cas9 and overexpression approaches allowed us to confirm the high specificity of our CAR construct for the tumor-associated CD44v6 as target antigen and to demonstrate a direct correlation between CD44v6 expression levels and cytotoxicity of the CAR T-cells. Importantly, the design of our clinically applicable lentiviral vector facilitates to co-express a second transgene for in vivo control of CAR T-cells, if undesired side-effects or toxicities occur.


Subject(s)
Head and Neck Neoplasms , Hyaluronan Receptors/immunology , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Squamous Cell Carcinoma of Head and Neck , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Humans , Hyaluronan Receptors/genetics , Neoplasm Recurrence, Local , Protein Isoforms , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/therapy , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
10.
PLoS One ; 16(4): e0250175, 2021.
Article in English | MEDLINE | ID: mdl-33891595

ABSTRACT

CD44 is a transmembrane glycoprotein that binds to hyaluronic acid, plays roles in a number of cellular processes and is expressed in a variety of cell types. It is up-regulated in stem cells and cancer. Anti-CD44 monoclonal antibodies affect cell motility and aggregation, and repress tumorigenesis and metastasis. Here we describe four new anti-CD44 monoclonal antibodies originating from B cells of a mouse injected with a plasmid expressing CD44 isoform 12. The four monoclonal antibodies bind to the terminal, extracellular, conserved domain of CD44 isoforms. Based on differences in western blot patterns of cancer cell lysates, the four anti-CD44 mAbs separated into three distinct categories that include P4G9, P3D2, and P3A7, and P3G4. Spot assay analysis with peptides generated in Escherichia coli support the conclusion that the monoclonal antibodies recognize unglycosylated sequences in the N-terminal conserved region between amino acid 21-220, and analyses with a peptide generated in human embryonic kidney 293 cells, demonstrate that these monoclonal antibodies bind to these peptides only after deglycosylation. Western blots with lysates from three cancer cell lines demonstrate that several CD44 isoforms are unglycosylated in the anti-CD44 target regions. The potential utility of the monoclonal antibodies in blocking tumorigenesis was tested by co-injection of cells of the breast cancer-derived tumorigenic cell line MDA-MB-231 with the anti-CD44 monoclonal antibody P3D2 into the mammary fat pads of mice. All five control mice injected with MDA-MB-231 cells plus anti-IgG formed palpable tumors, while only one of the six test mice injected with MDA-MB-231 cells plus P3D2 formed a tiny tumor, while the remaining five were tumor-free, indicating that the four anti-CD44 mAbs may be useful therapeutically.


Subject(s)
Antibodies, Monoclonal , Carcinogenesis/immunology , Hyaluronan Receptors/immunology , HEK293 Cells , Humans , MCF-7 Cells
11.
Int Immunopharmacol ; 95: 107502, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33690000

ABSTRACT

BACKGROUND: Immune thrombocytopenia (ITP) is an autoimmune hemorrhagic disease with a low platelet count. CD44 is a pivotal component involved in phagocytosis and inflammation, and monoclonal antibodies (mAbs) against CD44 have been shown to be beneficial in several autoimmune diseases. In the present study, we investigated the correlation between CD44 levels and disease severity in patients with ITP and explored the immunomodulatory mechanisms of the antihuman CD44 mAb BJ18 on platelet phagocytosis mediated by monocytes/macrophages. METHODS: Plasma was collected from 45 participants to measure the circulating concentration of CD44 using ELISA. Peripheral blood mononuclear cells from patients and controls were isolated and induced to differentiate into monocytes/macrophages utilizing cytokines and drugs. CD44 expression on circulating cells and the effects of BJ18 on platelet phagocytosis, Fcɣ receptor (FcɣR) expression and M1/M2 polarization of macrophages were evaluated using flow cytometry and qPCR. RESULTS: CD44 levels of both the soluble form found in plasma and the form expressed on the surface of circulating monocytes/macrophages were significantly elevated in ITP patients. Linear correlations were verified between the CD44 levels and major clinical characteristics. In an in vitro study, BJ18 successfully inhibited platelet phagocytosis by monocytes/macrophages obtained from ITP patients. Further studies indicated that BJ18 corrected abnormal FcγR expression on monocytes/macrophages. Moreover, the polarization of proinflammatory M1 macrophages could also be regulated by BJ18. CONCLUSIONS: Our data indicated that the CD44 level has potential predictive value for disease severity and that the antihuman CD44 mAb BJ18 may be a promising therapy for ITP patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Platelets , Hyaluronan Receptors/blood , Immunologic Factors/pharmacology , Macrophages/drug effects , Monocytes/drug effects , Purpura, Thrombocytopenic, Idiopathic/immunology , Receptors, IgG/immunology , Adolescent , Adult , Aged , Female , Humans , Hyaluronan Receptors/immunology , Macrophages/immunology , Male , Middle Aged , Monocytes/immunology , Phagocytosis/drug effects , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/genetics , Receptors, IgG/genetics , Young Adult
12.
Blood ; 137(15): 2114-2124, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33662988

ABSTRACT

Monoclonal immunoglobulin G (IgG) antibodies to CD44 (anti-CD44) are anti-inflammatory in numerous murine autoimmune models, but the mechanisms are poorly understood. Anti-CD44 anti-inflammatory activity shows complete therapeutic concordance with IV immunoglobulin (IVIg) in treating autoimmune disease models, making anti-CD44 a potential IVIg alternative. In murine immune thrombocytopenia (ITP), there is no mechanistic explanation for anti-CD44 activity, although anti-CD44 ameliorates disease similarly to IVIg. Here, we demonstrate a novel anti-inflammatory mechanism of anti-CD44 that explains disease amelioration by anti-CD44 in murine ITP. Macrophages treated with anti-CD44 in vitro had dramatically suppressed phagocytosis through FcγRs in 2 separate systems of IgG-opsonized platelets and erythrocytes. Phagocytosis inhibition by anti-CD44 was mediated by blockade of the FcγR IgG binding site without changing surface FcγR expression. Anti-CD44 of different subclasses revealed that FcγR blockade was specific to receptors that could be engaged by the respective anti-CD44 subclass, and Fc-deactivated anti-CD44 variants lost all FcγR-inhibiting activity. In vivo, anti-CD44 functioned analogously in the murine passive ITP model and protected mice from ITP when thrombocytopenia was induced through an FcγR that could be engaged by the CD44 antibody's subclass. Consistent with FcγR blockade, Fc-deactivated variants of anti-CD44 were completely unable to ameliorate ITP. Together, anti-CD44 inhibits macrophage FcγR function and ameliorates ITP consistent with an FcγR blockade mechanism. Anti-CD44 is a potential IVIg alternative and may be of particular benefit in ITP because of the significant role that FcγRs play in human ITP pathophysiology.


Subject(s)
Hyaluronan Receptors/immunology , Immunoglobulin G/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Receptors, IgG/immunology , Animals , Blood Platelets/immunology , Female , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Phagocytosis , RAW 264.7 Cells
13.
Exp Cell Res ; 402(1): 112522, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33771482

ABSTRACT

Immune escape is the main cause of the low response rate to immunotherapy for cancer, including ovarian cancer. Growth differentiation factor-15 (GDF-15) inhibits immune cell function. However, only few reports described the mechanism. Therefore, the aim of this study was to investigate the mechanism of immune escape regulated by GDF-15 in ovarian cancer. Ovarian cancer patients and healthy women were enrolled in this study. Immunohistochemistry and ELISA were performed to measure GDF-15 expression. Immunoprecipitation combined with mass spectrometry, surface plasmon resonance, and co-immunoprecipitation assay were used to evaluate the interaction between GDF-15 and the surface molecules of DCs. Immunofluorescence analysis, flow cytometry and transwell assay were used to evaluate additional effects of GDF-15 on DCs. The results showed that GDF-15 expression was higher in the ovarian cancer patients compared to that in the healthy women. The TIMER algorithm revealed that highly GDF-15 expression is associated with immune DC infiltration in immunoreactive high-grade serous carcinoma. A further study showed that GDF-15 suppressed DCs maturation, as well as IL-12p40 and TNF-α secretion, the length and number of protrusions and the migration. More importantly, CD44 in the surface of DCs interacted with GDF-15. The overexpression of CD44 in DCs resulted in the suppression of the inhibitory effect of GDF-15 on the length and number of DC synapses. In DCs overexpressing CD44 the inhibition of GDF-15 on the expression of CD11c, CD83 and CD86 was decreased, while in DCs with a knockdown of CD44 the inhibition was further enhanced. Knockdown of CD44 in DCs enhanced the inhibitory effect of GDF-15 on DC migration, while the overexpression of CD44 inhibited the inhibitory effect of GDF-15 on DC migration. In conclusion, the present study suggested that GDF-15 might facilitate ovarian cancer immune escape by interacting with CD44 in DCs to inhibit their function.


Subject(s)
Dendritic Cells/immunology , Growth Differentiation Factor 15/genetics , Hyaluronan Receptors/genetics , Ovarian Neoplasms/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Dendritic Cells/pathology , Female , Growth Differentiation Factor 15/immunology , Humans , Hyaluronan Receptors/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Middle Aged , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Th1 Cells/immunology , Tumor Escape/genetics , Tumor Escape/immunology
14.
Carbohydr Polym ; 251: 117103, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33142641

ABSTRACT

The polysaccharide-based biomaterials hyaluronic acid (HA) and chondroitin sulfate (CS) have aroused great interest for use in drug delivery systems for tumor therapy, as they have outstanding biocompatibility and great targeting ability for cluster determinant 44 (CD44). In addition, modified HA and CS can self-assemble into micelles or micellar nanoparticles (NPs) for targeted drug delivery. This review discusses the formation of HA- and CS-based NPs, and various types of CS-based NPs including CS-drug conjugates, CS-polymer NPs, CS-small molecule NPs, polyelectrolyte nanocomplexes (PECs), CS-metal NPs, and nanogels. We then focus on the applications of HA- and CS-based NPs in tumor chemotherapy, gene therapy, photothermal therapy (PTT), photodynamic therapy (PDT), sonodynamic therapy (SDT), and immunotherapy. Finally, this review is expected to provide guidelines for the development of various HA- and CS-based NPs used in multiple cancer therapies.


Subject(s)
Drug Delivery Systems/methods , Glycosaminoglycans/administration & dosage , Hyaluronan Receptors/immunology , Nanoparticles/administration & dosage , Neoplasms/drug therapy , Neoplasms/immunology , Animals , Chondroitin Sulfates/administration & dosage , Chondroitin Sulfates/chemistry , Clinical Trials as Topic , Doxorubicin/administration & dosage , Glycosaminoglycans/chemistry , Glycosaminoglycans/immunology , Humans , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/chemistry , Molecular Targeted Therapy , Nanoparticles/chemistry , Neoplasms/pathology , Neoplasms/therapy , Topoisomerase II Inhibitors/administration & dosage
15.
Curr Pharm Biotechnol ; 22(9): 1192-1205, 2021.
Article in English | MEDLINE | ID: mdl-33045964

ABSTRACT

BACKGROUND: The selection of a suitable signal peptide that can direct recombinant proteins from the cytoplasm to the extracellular space is an important criterion affecting the production of recombinant proteins in Escherichia coli, a widely used host. Nanobodies are currently attracting the attention of scientists as antibody alternatives due to their specific properties and feasibility of production in E. coli. OBJECTIVE: CD44 nanobodies constitute a potent therapeutic agent that can block CD44/HA interaction in cancer and inflammatory diseases. This molecule may also function as a drug against cancer cells and has been produced previously in E. coli without a signal peptide sequence. The goal of this project was to find a suitable signal peptide to direct CD44 nanobody extracellular secretion in E. coli that will potentially lead to optimization of experimental methods and facilitate downstream steps such as purification. METHODS: We analyzed 40 E. coli derived signal peptides retrieved from the Signal Peptide database and selected the best candidate signal peptides according to relevant criteria including signal peptide probability, stability, and physicochemical features, which were evaluated using signalP software version 4.1 and the ProtParam tool, respectively. RESULTS: In this in silico study, suitable candidate signal peptide(s) for CD44 nanobody secretory expression were identified. CSGA, TRBC, YTFQ, NIKA, and DGAL were selected as appropriate signal peptides with acceptable D-scores, and appropriate physicochemical and structural properties. Following further analysis, TRBC was selected as the best signal peptide to direct CD44 nanobody expression to the extracellular space of E. coli. CONCLUSION: The selected signal peptide, TRBC is the most suitable to promote high-level secretory production of CD44 nanobodies in E. coli and potentially will be useful for scaling up CD44 nanobody production in experimental research as well as in other CD44 nanobody applications. However, experimental work is needed to confirm the data.


Subject(s)
Escherichia coli/metabolism , Protein Sorting Signals/genetics , Single-Domain Antibodies/metabolism , Cloning, Molecular , Databases, Protein , Humans , Hyaluronan Receptors/immunology , Plasmids/genetics , Plasmids/metabolism , Recombinant Proteins/biosynthesis , Single-Domain Antibodies/genetics , Software
16.
Biochemistry (Mosc) ; 85(9): 1064-1081, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33050853

ABSTRACT

Tn antigen is a tumor-associated antigen that appears on cancer cells as a result of aberrant O-glycosylation. The most studied form of Tn antigen is found in mucins, in particular, in mucin 1 (MUC1). Antibodies against this form of Tn antigen are used to diagnose tumors, as well as to generate T-killers with a chimeric receptor. Some carcinomas do not carry MUC1 and antibodies of a different specificity are required to detect Tn antigen on these cells. In our work, we searched for anti-Tn antibodies without preliminary assumptions about the proteins that may be carriers of the Tn antigen. For this purpose, we obtained several pairs of isogenic cell lines with the wild type and knockout of the Cosmc gene, which is essential for correct protein O-glycosylation. Using the created lines as immunogens, we generated a monoclonal antibody AKC3, which reacted with the Cosmc-deficient A549 lung adenocarcinoma cells and did not bind to the wild-type cells. Using mass spectrometry, as well as co-immunoprecipitation, it was shown that the AKC3 antibody recognized the Tn antigen in the context of CD44 protein - a protein important for tumor growth. The AKC3 antibody can be used for tumor diagnosis, and to generate T cells with a chimeric receptor for treatment of tumors that do not express mucins.


Subject(s)
Adenocarcinoma of Lung/diagnosis , Antibodies, Monoclonal/immunology , Antigens, Tumor-Associated, Carbohydrate/metabolism , Biomarkers, Tumor/metabolism , Hyaluronan Receptors/metabolism , Lung Neoplasms/diagnosis , Molecular Chaperones/metabolism , A549 Cells , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/metabolism , Antigens, Tumor-Associated, Carbohydrate/immunology , CRISPR-Cas Systems , Glycosylation , Humans , Hyaluronan Receptors/immunology , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics
17.
Int J Oncol ; 57(4): 980-988, 2020 10.
Article in English | MEDLINE | ID: mdl-32945350

ABSTRACT

The impacts of post­operative abdominal infectious complications increase hematogenous distant metastasis and result in poor long­term survival after curative resection. Even if curative resection can be performed, the presence of circulating tumor cells is affected. The liver, the most common site of metastases, is an important organ in innate immune surveillance. However, the molecular mechanisms of distant hematogenous metastasis are not yet fully known. Platelets are crucial components in the tumor microenvironment that function to promote tumor progression and metastasis. The purpose of this study was to identify the effect of platelets on escape from innate immune surveillance in post­operative abdominal infectious complications. Platelet adherence was assessed by co­culturing human pancreatic cancer cells including transforming growth factor (TGF­ß)­treated BxPC­3. CD44 isoform, transcription factors and epithelial­mesenchymal transition markers were examined using western blotting. We also assessed whether cancer cells surrounded by activated platelets could escape from innate immune surveillance, using infectious and non­infectious mouse models injected intraperitoneally with LPS. Platelets were found to preferentially adhere to mesenchymal cells rather than epithelial cells. BxPC­3 epithelial cells showed upregulation of CD44­variant and epithelial splicing regulatory protein 1 (ESRP­1) expression. However, Panc­1 mesenchymal cells and TGF­ß­treated BxPC­3 cells showed upregulation of CD44­standard and zinc finger E­box­binding homeobox 1 (ZEB­1) expression and a reduction in ESRP­1. In the non­infectious model, cancer cells were not found in the liver. In the infectious model, although epithelial cells without platelet adhesion were in an apoptotic state, mesenchymal cells showed many viable cancer cells surrounded by activated platelets. Cancer cells were suggested to have phenotypic plasticity through the switching of CD44 isoforms. Mesenchymal cells, which express CD44­standard, could escape from immune surveillance by becoming surrounded by adhered activated platelets. Therefore, it may be necessary to administer antiplatelet agents to prevent distant hematogenous metastasis when post­operative abdominal infectious complications occur.


Subject(s)
Blood Platelets/immunology , Pancreatic Neoplasms/immunology , Platelet Adhesiveness/immunology , Tumor Microenvironment/immunology , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition , Humans , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Immunity, Innate , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Escape
18.
Cancer Res ; 80(19): 4185-4198, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32816856

ABSTRACT

Tumor-associated macrophages (TAM) in the tumor microenvironment (TME) cooperate with cancer stem cells (CSC) to maintain stemness. We recently identified cluster of differentiation 44 (CD44) as a surface marker defining head and neck squamous cell carcinoma (HNSCC) CSC. PI3K-4EBP1-SOX2 activation and signaling regulate CSC properties, yet the upstream molecular control of this pathway and the mechanisms underlying cross-talk between TAM and CSC in HNSCC remain largely unknown. Because CD44 is a molecular mediator in the TME, we propose here that TAM-influenced CD44 signaling could mediate stemness via the PI3K-4EBP1-SOX2 pathway, possibly by modulating availability of hyaluronic acid (HA), the main CD44 ligand. HNSCC IHC was used to identify TAM/CSC relationships, and in vitro coculture spheroid models and in vivo mouse models were used to identify the influence of TAMs on CSC function via CD44. Patient HNSCC-derived TAMs were positively and negatively associated with CSC marker expression at noninvasive and invasive edge regions, respectively. TAMs increased availability of HA and increased cancer cell invasion. HA binding to CD44 increased PI3K-4EBP1-SOX2 signaling and the CSC fraction, whereas CD44-VCAM-1 binding promoted invasive signaling by ezrin/PI3K. In vivo, targeting CD44 decreased PI3K-4EBP1-SOX2 signaling, tumor growth, and CSC. TAM depletion in syngeneic and humanized mouse models also diminished growth and CSC numbers. Finally, a CD44 isoform switch regulated epithelial-to-mesenchymal plasticity as standard form of CD44 and CD44v8-10 determined invasive and tumorigenic phenotypes, respectively. We have established a mechanistic link between TAMs and CSCs in HNSCC that is mediated by CD44 intracellular signaling in response to extracellular signals. SIGNIFICANCE: These findings establish a mechanistic link between tumor cell CD44, TAM, and CSC properties at the tumor-stroma interface that can serve as a vital area of focus for target and drug discovery.


Subject(s)
Head and Neck Neoplasms/pathology , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor-Associated Macrophages/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Feedback, Physiological , Female , Head and Neck Neoplasms/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Hyaluronic Acid/metabolism , Male , Mice, Inbred NOD , Monocytes/metabolism , Monocytes/pathology , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , SOXB1 Transcription Factors/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism , Xenograft Model Antitumor Assays , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
19.
Nanomedicine ; 29: 102257, 2020 10.
Article in English | MEDLINE | ID: mdl-32610072

ABSTRACT

Vaccine developmental strategies are utilizing antigens encapsulated in biodegradable polymeric nanoparticles. Here, we developed a Chlamydia nanovaccine (PLGA-rMOMP) by encapsulating its recombinant major outer membrane protein (rMOMP) in the extended-releasing and self-adjuvanting PLGA [poly (D, L-lactide-co-glycolide) (85:15)] nanoparticles. PLGA-rMOMP was small (nanometer size), round and smooth, thermally stable, and exhibited a sustained release of rMOMP. Stimulation of mouse primary dendritic cells (DCs) with PLGA-rMOMP augmented endosome processing, induced Th1 cytokines (IL-6 and IL-12p40), and expression of MHC-II and co-stimulatory (CD40, CD80, and CD86) molecules. BALB/c mice immunized with PLGA-rMOMP produced enhanced CD4+ T-cells-derived memory (CD44high CD62Lhigh), and effector (CD44high CD62Llow) phenotypes and functional antigen-specific serum IgG antibodies. In vivo biodistribution of PLGA-rMOMP revealed its localization within lymph nodes, suggesting migration from the injection site via DCs. Our data provide evidence that the PLGA (85:15) nanovaccine activates DCs and augments Chlamydia-specific rMOMP adaptive immune responses that are worthy of efficacy testing.


Subject(s)
Adaptive Immunity/genetics , Bacterial Outer Membrane Proteins/genetics , Nanoparticles/chemistry , Vaccines/immunology , Adaptive Immunity/immunology , Animals , Bacterial Outer Membrane Proteins/immunology , CD4 Antigens/chemistry , CD4 Antigens/immunology , Chlamydia/genetics , Chlamydia/immunology , Chlamydia/pathogenicity , Dendritic Cells/immunology , Histocompatibility Antigens Class II/genetics , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Interleukin-6/genetics , Interleukin-6/immunology , L-Selectin/chemistry , L-Selectin/immunology , Mice , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/immunology , T-Lymphocytes/immunology , Vaccines/genetics
20.
J Leukoc Biol ; 108(4): 1329-1337, 2020 10.
Article in English | MEDLINE | ID: mdl-32421902

ABSTRACT

CD8 T cells play a crucial role in immune responses to virus infections and tumors. Naïve CD8 T lymphocytes after TCR stimulation undergo differentiation into CTLs and memory cells, which are essential sources of IFN-γ. We investigated IFN-γ production by CD8 T cell subsets found in nonimmune mice. A minor fraction of in vitro TCR-stimulated CD8 T cells produce IFN-γ, and it is regulated at the transcriptional level. Antigen inexperienced C57BL/6 mice present the coexistence of 2 populations. The main population exhibits a CD44low CD122low profile, which is compatible with naïve lymphocytes. The minor expresses a phenotype of immunologic memory, CD44hi CD122hi . Both subsets are able to produce IL-2 in response to TCR activation, but only the memory-like population is responsible for IFN-γ production. Similar to memory CD8 T cells, CD44hi CD8+ T cells also present a higher level of the transcriptional factor Eomes and a lower level of T-bet (Tbx21) mRNA than CD44low CD8+ T cells. The presence of the CD44hi CD8+ T cell population in nonimmune OT-I transgenic mice reveals that the population is generated independently of antigenic stimulation. CpG methylation is an efficient epigenetic mechanism for gene silencing. DNA methylation at posttranscriptional CpG sites in the Ifng promoter is higher in CD44low CD8+ T cells than in CD44hi CD8+ T cells. Thus, memory-like CD8 T cells have a distinct epigenetic pattern in the Ifng promoter and can rapidly produce IFN-γ in response to TCR stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-gamma/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CpG Islands/immunology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Interferon-gamma/genetics , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...