Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Angiogenesis ; 21(3): 545-555, 2018 08.
Article in English | MEDLINE | ID: mdl-29500586

ABSTRACT

Galectin-9 consists of two peptide-linked carbohydrate recognition domains (CRDs), but alternative splicing and proteolytic processing can give rise to multiple galectin-9 isoforms. Some of these consist of a single CRD and can exert different functions in cell biology. Here, we explored the role of these galectin-9 isoforms in endothelial cell function and angiogenesis. For this, we compared the effects of the two separate CRDs (Gal-9N and Gal-9C) with the tandem repeat galectin-9M on endothelial cell proliferation, migration, sprouting and tube formation in vitro as well as on angiogenesis in vivo using the chicken chorioallantoic membrane (CAM) assay. Galectin-9 isoforms significantly affected proliferation in quiescent endothelial cells and migration in activated endothelial cells. Interestingly, both monovalent gal-9 CRDs displayed opposite effects compared to gal-9M on proliferation and migration. Sprouting was significantly inhibited by gal-9C, while all isoforms appeared to stimulate tube formation. Angiogenesis in vivo was hampered by all three isoforms with predominant effects on vessel length. In general, the isoforms induced only subtle concentration-dependent effects in vitro as well as in vivo. Collectively, the effects of different galectin-9 isoforms in endothelial cell biology depend on the cellular activation status. While opposing effects can be observed on a cellular level in vitro, all galectin-9 isoforms hamper angiogenesis in vivo. This warrants further investigation of the regulatory mechanisms that underlie the diverging roles of galectin-9 isoforms in endothelial cell biology since this could provide therapeutic opportunities.


Subject(s)
Cell Movement , Cell Proliferation , Galectins , Human Umbilical Vein Endothelial Cells , Neovascularization, Physiologic , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Chick Embryo , Chorioallantoic Membrane/anatomy & histology , Chorioallantoic Membrane/blood supply , Dose-Response Relationship, Drug , Galectins/chemistry , Galectins/metabolism , Galectins/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/pharmacology
2.
Haematologica ; 103(2): 325-335, 2018 02.
Article in English | MEDLINE | ID: mdl-29122991

ABSTRACT

Treatment of high-risk patients is a major challenge in multiple myeloma. This is especially true for patients assigned to the gene expression profiling-defined proliferation subgroup. Although recent efforts have identified some key players of proliferative myeloma, genetic interactions and players that can be targeted with clinically effective drugs have to be identified in order to overcome the poor prognosis of these patients. We therefore examined maternal embryonic leucine zipper kinase (MELK) for its implications in hyper-proliferative myeloma and analyzed the activity of the MELK inhibitor OTSSP167 both in vitro and in vivoMELK was found to be significantly overexpressed in the proliferative subgroup of myeloma. This finding translated into poor overall survival in patients with high vs low MELK expression. Enrichment analysis of upregulated genes in myeloma cells of MELKhigh patients confirmed the strong implications in myeloma cell proliferation. Targeting MELK with OTSSP167 impaired the growth and survival of myeloma cells, thereby affecting central survival factors such as MCL-1 and IRF4 This activity was also observed in the 5TGM.1 murine model of myeloma. OTSSP167 reduced bone marrow infiltration and serum paraprotein levels in a dose-dependent manner. In addition, we revealed a strong link between MELK and other proliferation-associated high-risk genes (PLK-1, EZH2, FOXM1, DEPDC1) and MELK inhibition also impaired the expression of those genes. We therefore conclude that MELK is an essential component of a proliferative gene signature and that pharmacological inhibition of MELK represents an attractive novel approach to overcome the poor prognosis of high-risk patients with a proliferative expression pattern.


Subject(s)
Cell Proliferation/drug effects , Multiple Myeloma/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Mice , Multiple Myeloma/pathology , Naphthyridines/pharmacology , Prognosis , Protein Serine-Threonine Kinases/metabolism , Risk Assessment
3.
Haematologica ; 103(8): 1359-1368, 2018 08.
Article in English | MEDLINE | ID: mdl-29748441

ABSTRACT

Multiple myeloma bone disease is characterized by an uncoupling of bone remodeling in the multiple myeloma microenvironment, resulting in the development of lytic bone lesions. Most myeloma patients suffer from these bone lesions, which not only cause morbidity but also negatively impact survival. The development of novel therapies, ideally with a combined anti-resorptive and bone-anabolic effect, is of great interest because lesions persist with the current standard of care, even in patients in complete remission. We have previously shown that MELK plays a central role in proliferation-associated high-risk multiple myeloma and its inhibition with OTSSP167 resulted in decreased tumor load. MELK inhibition in bone cells has not yet been explored, although some reports suggest that factors downstream of MELK stimulate osteoclast activity and inhibit osteoblast activity, which makes MELK inhibition a promising therapeutic approach. Therefore, we assessed the effect of OTSSP167 on bone cell activity and the development of myeloma-induced bone disease. OTSSP167 inhibited osteoclast activity in vitro by decreasing progenitor viability as well as via a direct anti-resorptive effect on mature osteoclasts. In addition, OTSSP167 stimulated matrix deposition and mineralization by osteoblasts in vitro This combined anti-resorptive and osteoblast-stimulating effect of OTSSP167 resulted in the complete prevention of lytic lesions and bone loss in myeloma-bearing mice. Immunohistomorphometric analyses corroborated our in vitro findings. In conclusion, we show that OTSSP167 has a direct effect on myeloma-induced bone disease in addition to its anti-multiple myeloma effect, which warrants further clinical development of MELK inhibition in multiple myeloma.


Subject(s)
Bone Diseases/drug therapy , Multiple Myeloma/drug therapy , Naphthyridines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Bone Diseases/etiology , Cell Line , Cell Proliferation/drug effects , Female , Heterografts , Humans , Mice , Mothers , Multiple Myeloma/complications , Multiple Myeloma/pathology , Naphthyridines/therapeutic use , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteolysis/drug therapy , Osteolysis/prevention & control , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
4.
Biochim Biophys Acta ; 1855(2): 235-47, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25819524

ABSTRACT

Galectins are a family of proteins that bind to specific glycans thereby deciphering the information captured within the glycome. In the last two decades, several galectin family members have emerged as versatile modulators of tumor progression. This has initiated the development and preclinical assessment of galectin-targeting compounds. With the first compounds now entering clinical trials it is pivotal to gain insight in the diagnostic and prognostic value of galectins in cancer as this will allow a more rational selection of the patients that might benefit most from galectin-targeted therapies. Here, we present a systematic review of galectin expression in human cancer patients. Malignant transformation is frequently associated with altered galectin expression, most notably of galectin-1 and galectin-3. In most cancers, increased galectin-1 expression is associated with poor prognosis while elevated galectin-9 expression is emerging as a marker of favorable disease outcome. The prognostic value of galectin-3 appears to be tumor type dependent and the other galectins require further investigation. Regarding the latter, additional studies using larger patient cohorts are essential to fully unravel the diagnostic and prognostic value of galectin expression. Furthermore, to better compare different findings, consensus should be reached on how to assess galectin expression, not only with regard to localization within the tissue and within cellular compartments but also regarding alternative splicing and genomic variations. Finally, linking galectin expression and function to aberrant glycosylation in cancer cells will improve our understanding of how these versatile proteins can be exploited for diagnostic, prognostic and even therapeutic purposes in cancer patients.


Subject(s)
Biomarkers, Tumor/biosynthesis , Galectin 1/biosynthesis , Galectin 3/biosynthesis , Neoplasms/genetics , Alternative Splicing/genetics , Biomarkers, Tumor/genetics , Blood Proteins , Cell Transformation, Neoplastic/genetics , Galectin 1/genetics , Galectin 3/genetics , Galectins , Gene Expression Regulation, Neoplastic , Genomics , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Prognosis
5.
Oncologist ; 21(3): 333-42, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26921288

ABSTRACT

Smoldering multiple myeloma (SMM) is an asymptomatic clonal plasma cell disorder and bridges monoclonal gammopathy of undetermined significance to multiple myeloma (MM), based on higher levels of circulating monoclonal immunoglobulin and bone marrow plasmocytosis without end-organ damage. Until a Spanish study reported fewer MM-related events and better overall survival among patients with high-risk SMM treated with lenalidomide and dexamethasone, prior studies had failed to show improved survival with earlier intervention, although a reduction in skeletal-related events (without any impact on disease progression) has been described with bisphosphonate use. Risk factors have now been defined, and a subset of ultra-high-risk patients have been reclassified by the International Myeloma Working Group as MM, and thus will require optimal MM treatment, based on biomarkers that identify patients with a >80% risk of progression. The number of these redefined patients is small (∼10%), but important to unravel, because their risk of progression to overt MM is substantial (≥80% within 2 years). Patients with a high-risk cytogenetic profile are not yet considered for early treatment, because groups are heterogeneous and risk factors other than cytogenetics are deemed to weight higher. Because patients with ultra-high-risk SMM are now considered as MM and may be treated as such, concerns exist that earlier therapy may increase the risk of selecting resistant clones and induce side effects and costs. Therefore, an even more accurate identification of patients who would benefit from interventions needs to be performed, and clinical judgment and careful discussion of pros and cons of treatment initiation need to be undertaken. For the majority of SMM patients, the standard of care remains observation until development of symptomatic MM occurs, encouraging participation in ongoing and upcoming SMM/early MM clinical trials, as well as consideration of bisphosphonate use in patients with early bone loss.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Dexamethasone/therapeutic use , Diphosphonates/therapeutic use , Multiple Myeloma/diagnosis , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Disease Progression , Europe , Humans , Immunoglobulin Light Chains/blood , Lenalidomide , Multiple Myeloma/pathology , Myeloma Proteins/analysis , Risk Factors , Thalidomide/therapeutic use , Tumor Burden
6.
Biochim Biophys Acta ; 1842(2): 284-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24333696

ABSTRACT

Galectins are carbohydrate binding proteins with versatile functions in tumor progression. Galectin-9, encoded by LGALS9, has been associated with metastasis and immunosuppression. We previously reported on regulation of LGALS9 expression during endothelial cell activation. Here, we show increased galectin-9 protein levels in the endothelium of different tumors, including carcinomas of the lung, liver, breast and kidney. Endothelial cells were found to express five LGALS9 splice variants, two of which have not been reported before. Splicing was found to be confined to exons 5, 6 and 10. Transfection of human microvascular endothelial cells (HMEC) with galectin-9∆5, a specific LGALS9 splice variant, induced a small but significant increase of proliferation, while migration was not affected by any LGALS9 splice variant. Application of recombinant galectin-9∆5 protein dose-dependently reduced proliferation and migration of HMEC as well as human umbilical vein endothelial cells in vitro. Enhanced sprouting and migration of human umbilical vein endothelial cell (HUVEC) towards a galectin-9∆5 gradient were observed. Interestingly, galectin-9∆5 was found to induce a small inhibitory effect on angiogenesis in vivo. Collectively, these data show that endothelial cells regulate the expression and splicing of LGALS9 during angiogenesis. The function of the dominant splice variant, i.e. galectin-9∆5, in endothelial cell biology depends on the concentration and environmental context in which it is presented to the cells.


Subject(s)
Alternative Splicing , Endothelial Cells/metabolism , Galectins/genetics , Gene Expression , Animals , Blotting, Western , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/physiology , Galectins/metabolism , Galectins/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Immunohistochemistry , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
7.
Biochim Biophys Acta ; 1836(1): 177-85, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23648450

ABSTRACT

Galectin family members have been shown to exert multiple roles in the context of tumor biology. Several recent findings support a similar multi-faceted role for galectin-9. Galectin-9 expression is frequently altered in cancer as compared to normal tissues. In addition, an increasing amount of evidence suggests that galectin-9 is involved in several aspects of tumor progression, including tumor cell adhesion and survival, immune escape and angiogenesis. Also, galectin-9 shows potential as a prognostic marker and a therapeutic target for several malignancies. In this review we summarize both the established and the emerging roles of galectin-9 in tumor biology and discuss the potential application of galectin-9 in anti-cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Galectins/metabolism , Neoplasms/pathology , Amino Acid Sequence , Animals , Galectins/antagonists & inhibitors , Humans , Mice , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/metabolism , Sequence Homology, Amino Acid
8.
Biol Reprod ; 88(1): 22, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23242525

ABSTRACT

Disruption of fetal-maternal tolerance mechanisms can contribute to pregnancy complications, including spontaneous abortion. Galectin-9 (LGALS9), a tandem repeat lectin associated with immune modulation, is expressed in the endometrium during the mid and late secretory phases and in decidua during human early pregnancy. However, the role of LGALS9 during pregnancy remains poorly understood. We used real-time PCR and immunohistochemical staining to analyze the expression of Lgals9/LGALS9 during mouse gestation as well as in human tissues obtained from normal pregnancy and spontaneous abortions. In mice, three Lgals9 splice variants were detected, the expression of which was differentially regulated during gestation. Furthermore, decidual Lgals9 expression was deregulated in a mouse model of spontaneous abortion, whereas placental levels did not change. We further found that the LGALS9 D5 isoform suppresses interferon gamma production by decidual natural killer cells. In human patients, six Lgals9 splice variants were detected, and a decrease in Lgals9 D5/10 was associated with spontaneous abortion. Altogether, these results show a differential regulation of Lgals9 isoform expression during normal and pathological pregnancies and designate Lgals9 as a potential marker for adverse pregnancy outcomes.


Subject(s)
Abortion, Spontaneous/metabolism , Galectins/metabolism , Gene Expression Regulation/physiology , Maternal-Fetal Relations/physiology , Animals , Biomarkers , Female , Galectins/genetics , Humans , Killer Cells, Natural , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Pregnancy , Protein Isoforms/genetics , Protein Isoforms/metabolism
9.
Biochim Biophys Acta ; 1805(1): 87-96, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19782719

ABSTRACT

Tumor angiogenesis facilitates tumor metastasis and allows malignant tissues to grow beyond a diffusion limited size. It is a complex process that requires endothelial cells to execute specific steps during different phases. miRNAs are small non-coding RNAs that act as molecular switches to redirect the expression profile of a cell. Evidence is emerging that miRNAs are important players in endothelial cell biology and tumor angiogenesis. In this review we summarize the available data of miRNA expression in the endothelium. In addition, we describe the current knowledge regarding the function of miRNAs in endothelial cell biology. Finally, we discuss the potential applications of miRNA based treatment strategies in angiostatic cancer therapy.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Humans
10.
Commun Biol ; 4(1): 1415, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34931005

ABSTRACT

Galectins are versatile glycan-binding proteins involved in immunomodulation. Evidence suggests that galectins can control the immunoregulatory function of cytokines and chemokines through direct binding. Here, we report on an inverse mechanism in which chemokines control the immunomodulatory functions of galectins. We show the existence of several specific galectin-chemokine binding pairs, including galectin-1/CXCL4. NMR analyses show that CXCL4 binding induces changes in the galectin-1 carbohydrate binding site. Consequently, CXCL4 alters the glycan-binding affinity and specificity of galectin-1. Regarding immunomodulation, CXCL4 significantly increases the apoptotic activity of galectin-1 on activated CD8+ T cells, while no effect is observed in CD4+ T cells. The opposite is found for another galectin-chemokine pair, i.e., galectin-9/CCL5. This heterodimer significantly reduces the galectin-9 induced apoptosis of CD4+ T cells and not of CD8+ T cells. Collectively, the current study describes an immunomodulatory mechanism in which specific galectin-chemokine interactions control the glycan-binding activity and immunoregulatory function of galectins.


Subject(s)
Chemokine CXCL5/metabolism , Galectin 1/metabolism , Galectins/metabolism , Immunomodulation , Platelet Factor 4/metabolism , Polysaccharides/metabolism , Humans , Jurkat Cells
11.
Adv Biol Regul ; 76: 100651, 2020 05.
Article in English | MEDLINE | ID: mdl-31519471

ABSTRACT

Opsismodysplasia (OPS) is a rare but severe autosomal recessive skeletal chondrodysplasia caused by inactivating mutations in the Inppl1/Ship2 gene. The molecular mechanism leading from Ship2 gene inactivation to OPS is currently unknown. Here, we used our Ship2Δ/Δ mouse expressing reduced amount of a catalytically-inactive SHIP2 protein and a previously reported SHIP2 inhibitor to investigate growth plate development and mineralization in vivo, ex vivo and in vitro. First, as observed in OPS patients, catalytic inactivation of SHIP2 in mouse leads to reduced body length, shortening of long bones, craniofacial dysmorphism, reduced height of the hyperthrophic chondrocyte zone and to defects in growth plate mineralization. Second, intrinsic Ship2Δ/Δ bone defects were sufficient to induce the characteristic OPS alterations in bone growth, histology and mineralization ex vivo. Third, expression of osteocalcin was significantly increased in SHIP2-inactivated chondrocyte cultures whereas production of mineralized nodules was markedly decreased. Targeting osteocalcin mRNA with a specific shRNA increased the production of mineralized nodules. Fourth, levels of p-MEK and p-Erk1/2 were significantly increased in SHIP2-inactivated chondrocytes in response to serum and IGF-1, but not to FGF2, as compared to control chondrocytes. Treatment of chondrocytes and bones in culture with a MEK inhibitor partially rescued the production of mineralized nodules, the size of the hypertrophic chondrocyte zone and bone growth, raising the possibility of a treatment that could partially reduce the phenotype of this severe condition. Altogether, our results indicate that Ship2Δ/Δ mice represent a relevant model for human OPS. They also highlight the important role of SHIP2 in chondrocytes during endochondral ossification and its different differentiation steps. Finally, we identified a role of osteocalcin in mineralized nodules production and for the MEK-Erk1/2 signaling pathway in the OPS phenotype.


Subject(s)
Chondrocytes/metabolism , MAP Kinase Kinase Kinases/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Osteocalcin/genetics , Osteochondrodysplasias/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Calcification, Physiologic/genetics , Cell Differentiation , Chondrocytes/pathology , Disease Models, Animal , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Growth Plate/metabolism , Growth Plate/pathology , Humans , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteocalcin/antagonists & inhibitors , Osteocalcin/metabolism , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/pathology , Osteogenesis/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/antagonists & inhibitors , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/deficiency , Phosphorylation/drug effects , Primary Cell Culture , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Thiophenes/pharmacology
12.
Cancers (Basel) ; 11(2)2019 Feb 23.
Article in English | MEDLINE | ID: mdl-30813402

ABSTRACT

Multiple myeloma osteolytic disease is caused by an uncoupled bone-remodelling process with an increased osteoclast activity. Disease development relies on interactions between myeloma cells and bone marrow stromal cells. Recent findings suggest a role for glycan-binding proteins in myeloma microenvironment. Here, we investigated lectins involved in osteoclastogenesis and their role in myeloma bone disease. Microarray data analysis showed a lower expression of galectin-1 (gal-1) in mature osteoclasts compared to monocytic progenitor cells, confirmed at the RNA and protein levels in osteoclast cultures. Confocal microscopy showed that gal-1 localised predominantly in the sealing zone of mature osteoclasts. Although equal differentiated-osteoclast numbers, gal-1-/- osteoclasts showed a higher resorption activity compared to wild-type controls. Micro-computed tomography showed an aberrant bone phenotype with decreased bone densities in gal-1-/- mice. In vivo, tumour progression was faster in gal-1-/- mice and associated with a marked bone loss. Additionally, myeloma cells were found to decrease gal-1 expression in osteoclasts. Our results demonstrate that galectin-1 regulates osteoclast activity with an increased resorption by gal-1-/- osteoclasts and decreased bone densities in gal-1-/- mice. We observed an enhanced tumour development in gal-1-/- mice compared to wild-type mice, suggesting that galectin-1 has a functional role in stromal cells in myeloma microenvironment.

13.
Leuk Lymphoma ; 59(1): 14-28, 2018 01.
Article in English | MEDLINE | ID: mdl-28573897

ABSTRACT

Multiple myeloma (MM) bone disease is a major cause of morbidity and mortality in MM patients and persists even in patients in remission. This bone disease is caused by an uncoupling of bone remodeling, with increased osteoclast and decreased osteoblast activity and formation, culminating in lytic bone destruction. Bisphosphonates are the current standard of care but new therapies are needed. As the molecular mechanisms controlling MM bone disease are increasingly well understood, new therapeutic targets are extensively explored in the preclinical setting and initial clinical trials with novel compounds now show promising results. In this review, we will provide a comprehensive overview of the biology of MM bone disease, summarize its current clinical management and discuss preclinical and clinical data on next generation therapies.


Subject(s)
Bone Diseases/etiology , Bone Diseases/therapy , Multiple Myeloma/complications , Animals , Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Bone Diseases/diagnosis , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Remodeling , Bone Resorption/drug therapy , Clinical Trials as Topic , Diphosphonates/pharmacology , Diphosphonates/therapeutic use , Disease Management , Drug Evaluation, Preclinical , Humans , Multiple Myeloma/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Signal Transduction , Treatment Outcome , Tumor Microenvironment
14.
Blood Cancer J ; 8(11): 105, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30409995

ABSTRACT

Progression of multiple myeloma (MM) is largely dependent on the bone marrow (BM) microenvironment wherein communication through different factors including extracellular vesicles takes place. This cross-talk not only leads to drug resistance but also to the development of osteolysis. Targeting vesicle secretion could therefore simultaneously ameliorate drug response and bone disease. In this paper, we examined the effects of MM exosomes on different aspects of osteolysis using the 5TGM1 murine model. We found that 5TGM1 sEVs, or 'exosomes', not only enhanced osteoclast activity, they also blocked osteoblast differentiation and functionality in vitro. Mechanistically, we could demonstrate that transfer of DKK-1 led to a reduction in Runx2, Osterix, and Collagen 1A1 in osteoblasts. In vivo, we uncovered that 5TGM1 exosomes could induce osteolysis in a similar pattern as the MM cells themselves. Blocking exosome secretion using the sphingomyelinase inhibitor GW4869 not only increased cortical bone volume, but also it sensitized the myeloma cells to bortezomib, leading to a strong anti-tumor response when GW4869 and bortezomib were combined. Altogether, our results indicate an important role for exosomes in the BM microenvironment and suggest a novel therapeutic target for anti-myeloma therapy.


Subject(s)
Bone Diseases/etiology , Bone Diseases/metabolism , Exosomes/metabolism , Multiple Myeloma/complications , Multiple Myeloma/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzylidene Compounds/pharmacology , Biomarkers , Bone Resorption/metabolism , Bortezomib/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Exosomes/ultrastructure , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Female , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/etiology , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteolysis , Standard of Care , Tumor Burden , Wnt Signaling Pathway
15.
Cancer Immunol Res ; 5(10): 839-846, 2017 10.
Article in English | MEDLINE | ID: mdl-28903971

ABSTRACT

Dysregulated expression of S100 protein family members is associated with cancer proliferation, invasion, angiogenesis, and inflammation. S100A9 induces myeloid-derived suppressor cell (MDSC) accumulation and activity. MDSCs, immunosuppressive cells that contribute to tumor immune escape, are the main producers of S100A9. In this study, we evaluated the role of extracellular S100A9 and the therapeutic relevance of S100A9 inhibition in multiple myeloma (MM), using the immunocompetent murine 5T33MM model. We demonstrated the presence of S100A9 and its receptor TLR4 in both monocytic and granulocytic MDSCs in human and mouse samples. We showed that S100A9 acted as a chemoattractant for MM cells and induced MDSCs to express and secrete inflammatory and pro-myeloma cytokines, including TNFα, IL6, and IL10. Blocking S100A9 interactions in vivo with the small molecule ABR-238901 did not directly affect MDSC accumulation but did reduce IL6 and IL10 cytokine expression by MDSC. ABR-238901 treatment in vivo reduced angiogenesis but had only minor effects on tumor load as single agent (6% reduction). However, ABR-238901 treatment in combination with bortezomib resulted in an increased reduction in tumor load compared with single treatments (50% relative reduction compared with bortezomib alone). Our data suggest that extracellular S100A9 promotes MM and that inhibition of S100A9 may have therapeutic benefit. Cancer Immunol Res; 5(10); 839-46. ©2017 AACR.


Subject(s)
Bone Marrow/metabolism , Bone Marrow/pathology , Calgranulin B/metabolism , Cytokines/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neovascularization, Pathologic/metabolism , Animals , Biomarkers , Bone Marrow Cells/metabolism , Cell Survival/genetics , Extracellular Space , Humans , Mice , Multiple Myeloma/genetics , Neovascularization, Pathologic/genetics
16.
Oncotarget ; 7(25): 37931-37943, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27177328

ABSTRACT

Multiple myeloma (MM) is a plasma cell malignancy characterized by the accumulation of tumor cells in the bone marrow (BM) and is associated with immunosuppression, angiogenesis and osteolysis. Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature, immunosuppressive myeloid cells that promote tumor progression through different mechanisms.In this work, we studied the contribution of MDSC subsets to different disease-promoting aspects in MM. We observed an expansion of polymorphonuclear/granulocytic (PMN-)MDSCs in two immunocompetent murine MM models, while this was not observed for monocytic (MO-)MDSCs. Both MDSC subpopulations from MM-bearing mice were immunosuppressive, but PMN-MDSCs displayed a higher suppressive potential. Soluble factors secreted by MM cells increased the viability of MDSCs, whereas the presence of MDSCs did not affect the proliferation of MM cells in vitro or in vivo. Interestingly, we observed a pro-angiogenic effect of PMN-MDSCs in the context of MM using the chick chorioallantoic membrane assay. Consistently, MM-derived PMN-MDSCs showed an up-regulation of angiogenesis-related factors and reduced PMN-MDSC levels were associated with less angiogenesis in vivo. Finally, we identified MO-MDSCs as osteoclast precursors.These results suggest that MDSC subpopulations play diverging roles in MM. We show for the first time that PMN-MDSCs exert a pro-angiogenic role in MM.


Subject(s)
Granulocytes/metabolism , Multiple Myeloma/genetics , Myeloid-Derived Suppressor Cells/metabolism , Neovascularization, Pathologic/metabolism , Animals , Humans , Mice , Multiple Myeloma/pathology , Neovascularization, Pathologic/pathology
17.
Oncotarget ; 7(21): 30712-29, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27095574

ABSTRACT

Multiple myeloma (MM)-associated osteolytic bone disease is a major cause of morbidity and mortality in MM patients and the development of new therapeutic strategies is of great interest. The proto-oncogene SRC is an attractive target for such a strategy. In the current study, we investigated the effect of treatment with the SRC inhibitor saracatinib (AZD0530) on osteoclast and osteoblast differentiation and function, and on the development of MM and its associated bone disease in the 5TGM.1 and 5T2MM murine MM models. In vitro data showed an inhibitory effect of saracatinib on osteoclast differentiation, polarization and resorptive function. In osteoblasts, collagen deposition and matrix mineralization were affected by saracatinib. MM cell proliferation and tumor burden remained unaltered following saracatinib treatment and we could not detect any synergistic effects with drugs that are part of standard care in MM. We observed a marked reduction of bone loss after treatment of MM-bearing mice with saracatinib as reflected by a restoration of trabecular bone parameters to levels observed in naive control mice. Histomorphometric analyses support that this occurs through an inhibition of bone resorption. In conclusion, these data further establish SRC inhibition as a promising therapeutic approach for the treatment of MM-associated osteolytic bone disease.


Subject(s)
Benzodioxoles/therapeutic use , Multiple Myeloma/drug therapy , Osteolysis/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogenes/drug effects , Quinazolines/therapeutic use , src-Family Kinases/antagonists & inhibitors , Administration, Oral , Animals , Bone and Bones/drug effects , Bone and Bones/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Myeloma/complications , Multiple Myeloma/pathology , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteolysis/etiology , Osteolysis/pathology , Proto-Oncogene Mas
18.
PLoS One ; 9(11): e113764, 2014.
Article in English | MEDLINE | ID: mdl-25415267

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is a malignant plasma cell disorder with poor long-term survival and high recurrence rates. Despite evidence of graft-versus-myeloma (GvM) effects, the use of allogeneic hematopoietic stem cell transplantation (allo-SCT) remains controversial in MM. In the current study, we investigated the anti-myeloma effects of allo-SCT from B10.D2 mice into MHC-matched myeloma-bearing Balb/cJ mice, with concomitant development of chronic graft-versus-host disease (GvHD). METHODS AND RESULTS: Balb/cJ mice were injected intravenously with luciferase-transfected MOPC315.BM cells, and received an allogeneic (B10.D2 donor) or autologous (Balb/cJ donor) transplant 30 days later. We observed a GvM effect in 94% of the allogeneic transplanted mice, as the luciferase signal completely disappeared after transplantation, whereas all the autologous transplanted mice showed myeloma progression. Lower serum paraprotein levels and lower myeloma infiltration in bone marrow and spleen in the allogeneic setting confirmed the observed GvM effect. In addition, the treated mice also displayed chronic GvHD symptoms. In vivo and in vitro data suggested the involvement of effector memory CD4 and CD8 T cells associated with the GvM response. The essential role of CD8 T cells was demonstrated in vivo where CD8 T-cell depletion of the graft resulted in reduced GvM effects. Finally, TCR Vß spectratyping analysis identified Vß families within CD4 and CD8 T cells, which were associated with both GvM effects and GvHD, whereas other Vß families within CD4 T cells were associated exclusively with either GvM or GvHD responses. CONCLUSIONS: We successfully established an immunocompetent murine model of graft-versus-myeloma. This is the first murine GvM model using immunocompetent mice that develop MM which closely resembles human MM disease and that are treated after disease establishment with an allo-SCT. Importantly, using TCR Vß spectratyping, we also demonstrated the presence of GvM unique responses potentially associated with the curative capacity of this immunotherapeutic approach.


Subject(s)
Graft vs Tumor Effect/immunology , Hematopoietic Stem Cell Transplantation , Multiple Myeloma/therapy , Neoplasms, Experimental/therapy , Allografts , Animals , Female , Male , Mice , Mice, Inbred BALB C , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
19.
PLoS One ; 9(9): e107988, 2014.
Article in English | MEDLINE | ID: mdl-25259711

ABSTRACT

Approximately 30-40% of the patients with early stage non-small cell lung cancer (NSCLC) will present with recurrent disease within two years of resection. Here, we performed extensive galectin expression profiling in a retrospective study using frozen and paraffin embedded tumor tissues from 87 stage I/II NSCLC patients. Our data show that galectin mRNA expression in NSCLC is confined to galectin-1, -3, -4, -7, -8, and -9. Next to stage, univariable Cox regression analysis identified galectin-1, galectin-9FL and galectin-9Δ5 as possible prognostic markers. Kaplan-Meier survival estimates revealed that overall survival was significantly shorter in patients that express galectin-1 above median levels, i.e., 23.0 (2.9-43.1) vs. 59.9 (47.7-72.1) months (p = 0.020) as well as in patients that express galectin-9Δ5 or galectin-9FL below the median, resp. 59.9 (41.9-75.9) vs. 32.8 (8.7-56.9) months (p = 0.014) or 23.2 (-0.4-46.8) vs. 58.9 (42.9-74.9) months (p = 0.042). All three galectins were also prognostic for disease free survival. Multivariable Cox regression analysis showed that for OS, the most significant prognostic model included stage, age, gal-1 and gal-9Δ5 while the model for DFS included stage, age and gal-9Δ5. In conclusion, the current study confirms the prognostic value of galectin-1 and identifies galectin-9Δ5 as novel potential prognostic markers in early stage NSCLC. These findings could help to identify early stage NSCLC patients that might benefit most from adjuvant chemotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Galectin 1/genetics , Galectins/genetics , Gene Expression Profiling , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Female , Galectin 1/metabolism , Galectins/metabolism , Gene Expression , Humans , Lung Neoplasms/mortality , Male , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Retrospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL