Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Transl Med ; 20(1): 380, 2022 08 29.
Article in English | MEDLINE | ID: mdl-36038904

ABSTRACT

BACKGROUND: Clarkson disease (monoclonal gammopathy-associated idiopathic systemic capillary leak syndrome, ISCLS) is a rare idiopathic condition marked by transient, relapsing-remitting episodes of systemic microvascular hyper-permeability, which liberates plasma fluid and macromolecules into the peripheral tissues. This pathology manifests clinically as the abrupt onset of hypotensive shock, hemoconcentration, and hypoalbuminemia. METHODS: We analysed endothelial glycocalyx (eGCX)-related markers in plasma from patients with ISCLS during acute disease flares and convalescence by ELISA and comprehensive proteomic profiling. We evaluated eGCX-related components and gene expression in cultured endothelial cells using RNA-sequencing, real-time PCR, and fluorescence staining. RESULTS: Serum levels of eGCX-related core components including hyaluronic acid (HA) and the core proteoglycan soluble syndecan-1 (sCD138) were elevated at baseline and during acute ISCLS flares. Serial measurements demonstrated that sCD138 levels peaked during the recovery (post-leak) phase of the illness. Proteomic analysis of matched acute and convalescent ISCLS plasma revealed increased abundance of eGCX-related proteins, including glypicans, thrombospondin-1 (TSP-1), and eGCX-degrading enzymes in acute compared to remission plasma. Abundance of endothelial cell damage markers did not differ in acute and baseline plasma. Expression of several eGCX-related genes and surface carbohydrate content in endothelial cells from patients with ISCLS did not differ significantly from that observed in healthy control cells. CONCLUSIONS: eGCX dysfunction, but not endothelial injury, may contribute to clinical symptoms of acute ISCLS. Serum levels of of eGCX components including sCD138 may be measured during acute episodes of ISCLS to monitor clinical status and therapeutic responses.


Subject(s)
Capillary Leak Syndrome , Biomarkers , Capillary Leak Syndrome/diagnosis , Capillary Leak Syndrome/pathology , Capillary Leak Syndrome/therapy , Endothelial Cells/pathology , Glycocalyx , Humans , Proteomics
2.
Curr Opin Clin Nutr Metab Care ; 25(1): 50-55, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34725313

ABSTRACT

PURPOSE OF REVIEW: Phosphatases of regenerating liver (PRL) are dual-specificity phosphatases and comprise three members, PRL-1, -2 and -3. Despite the importance of PRLs as oncoproteins, there is no consensus function for this family of phosphatases. In the current review paper, we summarize recent findings on the role of PRLs in metabolic regulation. RECENT FINDINGS: Reprogramming of cellular metabolism is a cancer hallmark. Glucose is the major source of energy in cells. Glucose metabolism occurs through the glycolysis and can continue through the pathways such as serine synthesis pathway or the tricarboxylic acid cycle (TCA). Magnesium (Mg2+), the second most abundant cation in cells, plays an essential role in energy production by acting as a cofactor for most enzymes involved in glycolysis and in TCA. Recent findings have shown that the PRL family has a role in metabolic reprogramming mediated by (1) Mg2+ homeostasis, (2) shifting the energy source preference to glucose consumption and fueling serine/glycine pathway and (3) regulating PI3 kinase/Mammalian target of rapamycin complex. Both the phosphatase and nonphosphatase activity of PRLs appear to be important for its oncogenic role. SUMMARY: The PRL family contributes to the metabolic plasticity of cancer cells and, thereby, allows cancer cells to meet the high metabolic demands required for cell proliferation.


Subject(s)
Neoplasms , Protein Tyrosine Phosphatases , Glycine , Humans , Liver , Serine
3.
FASEB J ; 35(3): e21344, 2021 03.
Article in English | MEDLINE | ID: mdl-33566385

ABSTRACT

Cancer cells often depend on microenvironment signals from molecules such as cytokines for proliferation and metabolic adaptations. PRL-3, a cytokine-induced oncogenic phosphatase, is highly expressed in multiple myeloma cells and associated with poor outcome in this cancer. We studied whether PRL-3 influences metabolism. Cells transduced to express PRL-3 had higher aerobic glycolytic rate, oxidative phosphorylation, and ATP production than the control cells. PRL-3 promoted glucose uptake and lactate excretion, enhanced the levels of proteins regulating glycolysis and enzymes in the serine/glycine synthesis pathway, a side branch of glycolysis. Moreover, mRNAs for these proteins correlated with PRL-3 expression in primary patient myeloma cells. Glycine decarboxylase (GLDC) was the most significantly induced metabolism gene. Forced GLDC downregulation partly counteracted PRL-3-induced aerobic glycolysis, indicating GLDC involvement in a PRL-3-driven Warburg effect. AMPK, HIF-1α, and c-Myc, important metabolic regulators in cancer cells, were not mediators of PRL-3's metabolic effects. A phosphatase-dead PRL-3 mutant, C104S, promoted many of the metabolic changes induced by wild-type PRL-3, arguing that important metabolic effects of PRL-3 are independent of its phosphatase activity. Through this study, PRL-3 emerges as one of the key mediators of metabolic adaptations in multiple myeloma.


Subject(s)
Multiple Myeloma/metabolism , Neoplasm Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Adenosine Triphosphate/biosynthesis , Cell Line, Tumor , Cell Proliferation , Glycine/metabolism , Glycine Dehydrogenase (Decarboxylating)/physiology , Glycolysis , Humans , Serine/metabolism
4.
Eur J Haematol ; 109(1): 31-40, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35276027

ABSTRACT

Cancer cells can convert proto-oncoproteins into oncoproteins by increasing the expression of genes that are oncogenic when expressed at high levels. Such genes can promote oncogenesis without being mutated. To find overexpressed genes in cancer cells from patients with multiple myeloma, we retrieved mRNA expression data from the CoMMpass database and ranked genes by their expression levels. We grouped the most highly expressed genes based on a set of criteria and we discuss the role a selection of them can play in the disease pathophysiology. The list was highly concordant with a similar list based on mRNA expression data from the PADIMAC study. Many well-known "myeloma genes" such as MCL1, CXCR4, TNFRSF17, SDC1, SLAMF7, PTP4A3, and XBP1 were identified as highly expressed, and we believe that hitherto unrecognized key players in myeloma pathogenesis are also enriched on the list. Highly expressed genes in malignant plasma cells that were absent or expressed at only a low level in healthy plasma cells included IFI6, IFITM1, PTP4A3, SIK1, ALDOA, ATP5MF, ATP5ME, and PSMB4. The ambition of this article is not to validate the role of each gene but to serve as a guide for studies aiming at identifying promising treatment targets.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Oncogenes , Plasma Cells/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/metabolism
5.
Molecules ; 26(24)2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34946532

ABSTRACT

Cytosolic phospholipase A2α (cPLA2α) is the rate-limiting enzyme in releasing arachidonic acid and biosynthesis of its derivative eicosanoids. Thus, the catalytic activity of cPLA2α plays an important role in cellular metabolism in healthy as well as cancer cells. There is mounting evidence suggesting that cPLA2α is an interesting target for cancer treatment; however, it is unclear which cancers are most relevant for further investigation. Here we report the relative expression of cPLA2α in a variety of cancers and cancer cell lines using publicly available datasets. The profiling of a panel of cancer cell lines representing different tissue origins suggests that hematological malignancies are particularly sensitive to the growth inhibitory effect of cPLA2α inhibition. Several hematological cancers and cancer cell lines overexpressed cPLA2α, including multiple myeloma. Multiple myeloma is an incurable hematological cancer of plasma cells in the bone marrow with an emerging requirement of therapeutic approaches. We show here that two cPLA2α inhibitors AVX420 and AVX002, significantly and dose-dependently reduced the viability of multiple myeloma cells and induced apoptosis in vitro. Our findings implicate cPLA2α activity in the survival of multiple myeloma cells and support further studies into cPLA2α as a potential target for treating hematological cancers, including multiple myeloma.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acids, Omega-3/pharmacology , Group IV Phospholipases A2 , Multiple Myeloma , Neoplasm Proteins , Cell Line, Tumor , Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/metabolism , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism
6.
Haematologica ; 102(7): 1266-1272, 2017 07.
Article in English | MEDLINE | ID: mdl-28385781

ABSTRACT

Circulating tumor DNA is a promising biomarker to monitor tumor load and genome alterations. We explored the presence of circulating tumor DNA in multiple myeloma patients and its relation to disease activity during long-term follow-up. We used digital droplet polymerase chain reaction analysis to monitor recurrent mutations, mainly in mitogen activated protein kinase pathway genes NRAS, KRAS and BRAF Mutations were identified by next-generation sequencing or polymerase chain reaction analysis of bone marrow plasma cells, and their presence analyzed in 251 archived serum samples obtained from 20 patients during a period of up to 7 years. In 17 of 18 patients, mutations identified in bone marrow during active disease were also found in a time-matched serum sample. The concentration of mutated alleles in serum correlated with the fraction in bone marrow plasma cells (r=0.507, n=34, P<0.002). There was a striking covariation between circulating mutation levels and M protein in ten out of 11 patients with sequential samples. When relapse evaluation by circulating tumor DNA and M protein could be directly compared, the circulating tumor DNA showed relapse earlier in two patients (3 and 9 months), later in one patient (4 months) and in three patients there was no difference. In three patients with transformation to aggressive disease, the concentrations of mutations in serum increased up to 400 times, an increase that was not seen for the M protein. In conclusion, circulating tumor DNA in myeloma is a multi-faceted biomarker reflecting mutated cells, total tumor mass and transformation to a more aggressive disease. Its properties are both similar and complementary to M protein.


Subject(s)
Biomarkers, Tumor , Cell-Free Nucleic Acids , DNA, Neoplasm , Multiple Myeloma/genetics , Mutation , Aged , Biomarkers , DNA Mutational Analysis , Disease Progression , Female , Humans , Liquid Biopsy , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Multiple Myeloma/therapy , Myeloma Proteins , Neoplasm Staging , Retrospective Studies , Exome Sequencing
7.
Genes Chromosomes Cancer ; 55(11): 890-901, 2016 11.
Article in English | MEDLINE | ID: mdl-27311012

ABSTRACT

Multiple myeloma can be divided into two distinct genetic subgroups: hyperdiploid (HRD) or nonhyperdiploid (NHRD) myeloma. Myeloma cell lines are important tools to study myeloma cell biology and are commonly used for preclinical screening and testing of new drugs. With few exceptions human myeloma cell lines are derived from NHRD patients, even though about half of the patients have HRD myeloma. Thus, there is a need for cell lines of HRD origin to enable more representative preclinical studies. Here, we present two novel myeloma cell lines, VOLIN and KJON. Both of them were derived from patients with HRD disease and shared the same genotype as their corresponding primary tumors. The cell lines' chromosomal content, genetic aberrations, gene expression, immunophenotype as well as some of their growth characteristics are described. Neither of the cell lines was found to harbor immunoglobulin heavy chain translocations. The VOLIN cell line was established from a bone marrow aspirate and KJON from peripheral blood. We propose that these unique cell lines may be used as tools to increase our understanding of myeloma cell biology. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Line, Tumor , Multiple Myeloma/pathology , Aneuploidy , Diploidy , Humans , Immunophenotyping , Multiple Myeloma/genetics , Translocation, Genetic
8.
J Transl Med ; 14: 71, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26975394

ABSTRACT

BACKGROUND: PRL-3 is a phosphatase implicated in oncogenesis in multiple cancers. In some cancers, notably carcinomas, PRL-3 is also associated with inferior prognosis and increased metastatic potential. In this study we investigated the expression of PRL-3 mRNA in fresh-frozen samples from patients undergoing radical prostatectomy because of prostate cancer (PC) and the biological function of PRL-3 in prostate cancer cells. METHODS: Samples from 41 radical prostatectomy specimens (168 samples in total) divided into low (Gleason score ≤ 6), intermediate (Gleason score = 7) and high (Gleason score ≥ 8) risk were analyzed with gene expression profiling and compared to normal prostate tissue. PRL-3 was identified as a gene with differential expression between healthy and cancerous tissue in these analyses. We used the prostate cancer cell lines PC3 and DU145 and a small molecular inhibitor of PRL-3 to investigate whether PRL-3 had a functional role in cancer. Relative ATP-measurement and thymidine incorporation were used to assess the effect of PRL-3 on growth of the cancer cells. We performed an in vitro scratch assay to investigate the involvement of PRL-3 in migration. Immunohistochemistry was used to identify PRL-3 protein in prostate cancer primary tumor and corresponding lymph node metastases. RESULTS: Compared to normal prostate tissue, the prostate cancer tissue expressed a significantly higher level of PRL-3. We found PRL-3 to be present in both PC3 and DU145, and that inhibition of PRL-3 led to growth arrest and apoptosis in these two cell lines. Inhibition of PRL-3 led to reduced migration of the PC3 cells. Immunohistochemistry showed PRL-3 expression in both primary tumor and corresponding lymph node metastases. CONCLUSIONS: PRL-3 mRNA was expressed to a greater extent in prostate cancer tissue compared to normal prostate tissue. PRL-3 protein was expressed in both prostate cancer primary tumor and corresponding lymph node metastases. The results from our in vitro assays suggest that PRL-3 promotes growth and migration in prostate cancer. In conclusion, these results imply that PRL-3 has a role in the pathogenesis of prostate cancer.


Subject(s)
Cell Movement , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Tyrosine Phosphatases/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell Survival , Genetic Loci , Humans , Lymphatic Metastasis , Male , Neoplasm Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Tissue Array Analysis
9.
Genes Chromosomes Cancer ; 54(11): 692-701, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26220195

ABSTRACT

The growth and survival factor hepatocyte growth factor (HGF) is expressed at high levels in multiple myeloma (MM) cells. We report here that elevated HGF transcription in MM was traced to DNA mutations in the promoter alleles of HGF. Sequence analysis revealed a previously undiscovered single-nucleotide polymorphism (SNP) and crucial single-nucleotide variants (SNVs) in the promoters of myeloma cells that produce large amounts of HGF. The allele-specific mutations functionally reassembled wild-type sequences into the motifs that affiliate with endogenous transcription factors NFKB (nuclear factor kappa-B), MZF1 (myeloid zinc finger 1), and NRF-2 (nuclear factor erythroid 2-related factor 2). In vitro, a mutant allele that gained novel NFKB-binding sites directly responded to transcriptional signaling induced by tumor necrosis factor alpha (TNFα) to promote high levels of luciferase reporter. Given the recent discovery by genome-wide sequencing (GWS) of numerous non-coding mutations in myeloma genomes, our data provide evidence that heterogeneous SNVs in the gene regulatory regions may frequently transform wild-type alleles into novel transcription factor binding properties to aberrantly interact with dysregulated transcriptional signals in MM and other cancer cells.


Subject(s)
Alleles , Gene Expression Regulation, Neoplastic , Genome, Human , Multiple Myeloma/genetics , Mutation , Transcription Factors/metabolism , Binding Sites , Cell Line, Tumor , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Multiple Myeloma/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic
10.
Mediators Inflamm ; 2014: 438070, 2014.
Article in English | MEDLINE | ID: mdl-24882950

ABSTRACT

In sarcoidosis, increased Th17 cell fractions have been reported in bronchoalveolar lavage fluid, and elevated numbers of Th17 cells producing IFN- γ have been observed in peripheral blood. The balance between Th1, Th17, and FoxP3(+) CD4(+) T cell subsets in sarcoidosis remains unclear. Bronchoalveolar lavage fluid cells, from 30 patients with sarcoidosis, 18 patients with other diffuse parenchymal lung diseases, and 15 healthy controls, were investigated with flow cytometry for intracellular expression of FoxP3. In a subset of the patients, expression of the cytokines IL17A and IFN- γ was investigated. The fractions of FoxP3(+) CD4(+) T cells and Th17 cells were both lower in sarcoidosis compared to controls (P = 0.017 and P = 0.011, resp.). The proportion of Th17 cells positive for IFN- γ was greater in sarcoidosis than controls (median 72.4% versus 31%, P = 0.0005) and increased with radiologic stage (N = 23, rho = 0.45, and P = 0.03). IFN- γ (+) Th17 cells were highly correlated with Th1 cells (N = 23, rho = 0.64, and P = 0.001), and the ratio of IFN- γ (+) Th17/FoxP3(+) CD4(+) T cells was prominently increased in sarcoidosis. IFN- γ (+) Th17 cells may represent a pathogenic subset of Th17 cells, yet their expression of IFN- γ could be a consequence of a Th1-polarized cytokine milieu. Our results indicate a possible immune cell imbalance in sarcoidosis.


Subject(s)
Bronchoalveolar Lavage Fluid , Interferon-gamma/metabolism , Sarcoidosis, Pulmonary/immunology , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology , Adult , Aged , CD4-Positive T-Lymphocytes/cytology , Case-Control Studies , Cytokines/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Humans , Lung Diseases/metabolism , Male , Middle Aged , Phenotype , Sarcoidosis/immunology , Th1 Cells/cytology
12.
Eur J Haematol ; 91(3): 196-200, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23607294

ABSTRACT

OBJECTIVES: Decorin is a stromal-produced small leucine-rich proteoglycan known to attenuate tumour pro-survival, migration, proliferation and angiogenic signalling pathways. Recent studies have shown that decorin interacts with the hepatocyte growth factor (HGF) receptor c-Met, a potential key pathway in multiple myeloma (MM). METHODS: Decorin levels in paired peripheral blood and bone marrow plasma samples from healthy volunteers (HV) (n = 23), and patients with monoclonal gammopathy of undetermined significance (MGUS) (n = 41) and MM (n = 19) were determined by ELISA. Further, the ability of decorin to inhibit HGF-induced effects on MM cell lines were analysed in vitro using cell viability and Transwell migration assays. RESULTS: We found that decorin concentrations were significantly higher (P < 0.05) in bone marrow (BM) plasma from HVs (median 35.2 ng/mL; range, 15.3-99.1) compared with MGUS (median 22.5 ng/mL; range, 11.1-59.5) and patients with MM (median 21.5 ng/mL; range, 10.6-35.9). Decorin levels were higher in BM plasma than in peripheral blood in all groups, with a BM/PB ratio of 3.9, 3.4 and 2.5 for HV, MGUS and MM, respectively. A positive correlation (Spearman's ρ = 0.51, P < 0.05) was found between simultaneously measured levels of HGF and decorin in BM plasma in HVs, but not in MGUS or MM samples. Functionally, decorin inhibited HGF-induced migration and viability of INA-6 and ANBL-6 MM cell lines, independent of c-Met down-regulation. CONCLUSION: Our results show that decorin is down-regulated in MGUS and MM bone marrow plasma and that it inhibits HGF-induced viability and migration of myeloma cell lines in vitro.


Subject(s)
Bone Marrow Cells/metabolism , Decorin/metabolism , Monoclonal Gammopathy of Undetermined Significance/metabolism , Multiple Myeloma/metabolism , Plasma Cells/metabolism , Aged , Aged, 80 and over , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Down-Regulation , Female , Hepatocyte Growth Factor/pharmacology , Humans , Male , Middle Aged , Plasma Cells/drug effects , Plasma Cells/pathology , Proto-Oncogene Proteins c-met/metabolism
13.
Eur J Haematol ; 91(5): 399-410, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23952536

ABSTRACT

BACKGROUND: c-MET is the tyrosine kinase receptor of the hepatocyte growth factor (HGF). HGF-c-MET signaling is involved in many human malignancies, including multiple myeloma (MM). Recently, multiple agents have been developed directed to interfere at different levels in HGF-c-MET signaling pathway. Nanobodies are therapeutic proteins based on the smallest functional fragments of heavy-chain-only antibodies. In this study, we wanted to determine the anticancer effect of a novel anti-c-MET Nanobody in MM. METHODS: We examined the effects of an anti-c-MET Nanobody on thymidine incorporation, migration, adhesion of MM cells, and osteoblastogenesis in vitro. Furthermore, we investigated the effects of the Nanobody on HGF-dependent c-MET signaling by Western blotting. RESULTS: We show that the anti-c-MET Nanobody effectively inhibited thymidine incorporation of ANBL-6 MM cells via inhibition of an HGF autocrine growth loop and thymidine incorporation in INA-6 MM cells induced by exogenous HGF. HGF-induced migration and adhesion of INA-6 were completely and specifically blocked by the Nanobody. Furthermore, the Nanobody abolished the inhibiting effect of HGF on bone morphogenetic protein-2-induced alkaline phosphatase activity and the mineralization of human mesenchymal stem cells. Finally, we show that the Nanobody reduced phosphorylation of tyrosine residues in c-MET, MAPK, and Akt. We also compared the Nanobody with anti-c-MET monoclonal antibodies and revealed the similar or better effect. CONCLUSIONS: The anti-c-MET Nanobody inhibited MM cell migration, thymidine incorporation, and adhesion, and blocked the HGF-mediated inhibition of osteoblastogenesis. The anti-c-MET Nanobody might represent a novel therapeutic agent in the treatment of MM and other cancers driven by HGF-c-MET signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Signal Transduction/drug effects , Single-Domain Antibodies/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Hepatocyte Growth Factor/pharmacology , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteogenesis/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/genetics , Thymidine/metabolism
14.
Eur J Immunol ; 41(2): 437-49, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268013

ABSTRACT

Serglycin (SG) is a proteoglycan expressed by hematopoietic cells and is constitutively secreted by multiple myeloma (MM) cells. SG participates in the regulation of various inflammatory events. We found that SG secreted by human MM cell lines inhibits both the classical and lectin pathways of complement, without influencing alternative pathway activity. The inhibitory effect of SG is due to direct interactions with C1q and mannose-binding lectin (MBL). C1q-binding is mediated through the glycosaminoglycan moieties of SG, whereas MBL binds additionally to SG protein core. Interactions between SG and C1q as well as MBL are diminished in the presence of chondroitin sulfate type E. In addition, we localized the SG-binding site to the collagen-like stalk of C1q. Interactions between SG and C1q as well as MBL are ionic in character and only the interaction with MBL was found to be partially dependent on the presence of calcium. We found the serum levels of SG to be elevated in patients with MM compared to healthy controls. Moreover, we found that SG expressed from myeloma plasma cells protects these cells from complement activation induced by treatment with anti-thymocyte immunoglobulins. This might protect myeloma cells during immunotherapy and promote survival of malignant cells.


Subject(s)
Complement Pathway, Classical/drug effects , Complement Pathway, Mannose-Binding Lectin/drug effects , Glycosaminoglycans/metabolism , Multiple Myeloma/immunology , Proteoglycans/pharmacology , Vesicular Transport Proteins/pharmacology , Aged , Animals , Antibodies, Neoplasm/immunology , Binding Sites/immunology , Cell Line, Tumor , Complement C1q/metabolism , Complement C3b/metabolism , Complement C4b/metabolism , Complement Pathway, Alternative/drug effects , Complement Pathway, Classical/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , Complement System Proteins/metabolism , Female , Glycosaminoglycans/pharmacology , Hemolysis/drug effects , Hemolysis/immunology , Humans , Male , Mannose-Binding Lectin/metabolism , Middle Aged , Multiple Myeloma/blood , Protein Binding/drug effects , Protein Binding/immunology , Proteoglycans/blood , Proteoglycans/immunology , Proteoglycans/metabolism , Rabbits , Sheep , Vesicular Transport Proteins/blood , Vesicular Transport Proteins/immunology , Vesicular Transport Proteins/metabolism
15.
Histopathology ; 60(3): 443-51, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22276607

ABSTRACT

AIMS: Interaction with the bone marrow microenvironment is important for homing and survival of myeloma cells. One cytokine involved in this process is hepatocyte growth factor (HGF). HGF, by binding to the receptor tyrosine kinase c-Met, mediates a broad range of tumour progression activities. Our aims were to investigate whether HGF and c-Met are present in bone marrow and extramedullary tumours from patients with monoclonal plasma cell disease, and whether c-Met is activated. METHODS AND RESULTS: Expression of HGF, c-Met and phospho-c-Met was studied by immunohistochemistry in biopsies from 80 patients with monoclonal plasma cell disease. Cytoplasmic staining for HGF in plasma cells was demonstrated in 58 of 68 biopsies from multiple myeloma patients (85%), but also in biopsies from nine of 10 healthy individuals. Membranous staining for c-Met was found in 25 of 63 multiple myeloma patients (40%) and in none of 10 healthy individuals. Membranous staining for phospho-c-Met was found in biopsies from 15 of 21 c-Met-positive myeloma patients (71%). CONCLUSIONS: Our data point to c-Met expression as one of the factors that distinguishes normal from malignant plasma cells, and indicate that the HGF/c-Met system is activated in multiple myeloma patients.


Subject(s)
Bone Marrow Neoplasms/diagnosis , Hepatocyte Growth Factor/metabolism , Multiple Myeloma/diagnosis , Plasma Cells/pathology , Proto-Oncogene Proteins c-met/metabolism , Adult , Aged , Aged, 80 and over , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Neoplasms/classification , Bone Marrow Neoplasms/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Cytoplasm/metabolism , Cytoplasm/pathology , Disease Progression , Female , Humans , Male , Middle Aged , Multiple Myeloma/classification , Multiple Myeloma/metabolism , Phosphorylation , Plasma Cells/metabolism
16.
Oncotarget ; 13: 1175-1186, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36268559

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is an incurable malignancy of plasma cells. The serine protease matriptase is frequently dysregulated in human carcinomas, which facilitates tumor progression and metastatic dissemination. The importance of matriptase in hematological malignancies is yet to be clarified. In this study, we aimed to characterize the role of matriptase in MM. MATERIALS AND METHODS: mRNA expression of matriptase and its inhibitors hepatocyte growth factor activator inhibitor (HAI)-1 and HAI-2 was studied in primary MM cells from patient samples and human myeloma cell lines (HMCLs). We further investigated the effect of matriptase on migration and proliferation of myeloma cells in vitro. By use of the CoMMpass database, we assessed the clinical relevance of matriptase in MM patients. RESULTS: Matriptase was expressed in 96% of patient samples and all HMCLs tested. Overexpression of matriptase in vitro reduced proliferation, and significantly decreased cytokine-induced migration. Conversely, matriptase knockdown significantly enhanced migration. Mechanistically, overexpression of matriptase inhibited activation of Src kinase. CONCLUSIONS: Our findings may suggest a novel role of matriptase as a tumor suppressor in MM pathogenesis.


Subject(s)
Multiple Myeloma , Humans , Proteinase Inhibitory Proteins, Secretory/metabolism , Multiple Myeloma/genetics , Serine Proteases , RNA, Messenger/metabolism , src-Family Kinases , Cytokines , Cell Proliferation
17.
Eur J Haematol ; 87(5): 394-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21466586

ABSTRACT

OBJECTIVES: The receptor tyrosine kinase c-Met and its ligand, hepatocyte growth factor (HGF), play key roles in tumour genesis and metastasis and contribute in multiple myeloma pathogenesis. Substantial data support that a soluble extracellular fragment of c-Met may function as a decoy receptor that downregulates the biological effects of HGF and c-Met. We examined serum levels of soluble c-Met in patients with myeloma and healthy individuals and investigated a possible relationship with clinical disease parameters and survival. METHODS: The concentration of c-Met and HGF was measured by enzyme-linked immunosorbent assay in serum (n=49) and bone marrow plasma (n=16) from patients with multiple myeloma and in serum from healthy controls (n=26). RESULTS: The median serum concentration of soluble c-Met was 186 ng/mL (range 22-562) in patients with multiple myeloma and 189 ng/mL (range 124-397) in healthy individuals. There was a significant negative correlation between serum c-Met levels and disease stage, bone marrow plasma cell percentage and serum concentration of M-protein. CONCLUSION: We have for the first time examined the concentration of soluble c-Met in serum from patients with myeloma and found equal median levels in patients with myeloma as a group and healthy individuals. Still, serum levels of soluble c-Met correlated negatively with parameters of disease burden in patients with myeloma. We suggest that a possible role for the c-Met ectodomain as a negative regulator of HGF/c-Met activity should be examined in multiple myeloma.


Subject(s)
Biomarkers, Tumor/blood , Multiple Myeloma/blood , Proto-Oncogene Proteins c-met/blood , Female , Humans , Male
18.
Cancer Lett ; 501: 105-113, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33290866

ABSTRACT

Many cell signaling pathways are activated or deactivated by protein tyrosine phosphorylation and dephosphorylation, catalyzed by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs), respectively. Even though PTPs are as important as PTKs in this process, their role has been neglected for a long time. Multiple myeloma (MM) is a cancer of plasma cells, which is characterized by production of monoclonal immunoglobulin, anemia and destruction of bone. MM is still incurable with high relapse frequency after treatment. In this review, we highlight the PTPs that were previously described in MM or have a role that can be relevant in a myeloma context. Our purpose is to show that despite the importance of PTPs in MM pathogenesis, many unanswered questions in this field need to be addressed. This might help to detect novel treatment strategies for MM patients.


Subject(s)
Multiple Myeloma/enzymology , Protein Tyrosine Phosphatases/metabolism , Animals , Humans
19.
Cancers (Basel) ; 13(8)2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33918618

ABSTRACT

Non-small cell lung carcinoma (NSCLC) is one of the most commonly diagnosed cancers and a leading cause of cancer-related deaths. Immunotherapy with immune checkpoint inhibitors shows beneficial responses, but only in a proportion of patients. To improve immunotherapy in NSCLC, we need to map the immune checkpoints that contribute immunosuppression in NSCLC-associated immune cells and to identify novel pathways that regulate immunosuppression. Here, we investigated the gene expression profiles of intra-tumoral immune cells isolated from NSCLC patients and compared them to the expression profiles of their counterparts in adjacent healthy tissue. Transcriptome analysis was performed on macrophages, CD4+ and CD8+ T cells. The data was subjected to Gene Ontology (GO) term enrichment and weighted correlation network analysis in order to identify mediators of immunosuppression in the tumor microenvironment in NSCLC. Immune cells from NSCLC revealed a consistent differential expression of genes involved in interactions between myeloid cells and lymphocytes. We further identified several immunosuppressive molecules and pathways that may be activated in tumor-associated macrophages in NSCLC. Importantly, we report novel data on immune cell expression of the newly described CD200/CD200R1 pathway, and the leukocyte immunoglobulin-like receptors (LILRs), which may represent novel innate immune checkpoints, dampening the anti-tumor T cell immune response in NSCLC. Our study substantiates the importance of tumor-associated macrophages as a mediator of immunosuppression and a promising target for immunotherapy.

SELECTION OF CITATIONS
SEARCH DETAIL