Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Blood ; 139(4): 608-623, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34657151

ABSTRACT

The key immunologic signatures associated with clinical outcomes after posttransplant cyclophosphamide (PTCy)-based HLA-haploidentical (haplo) and HLA-matched bone marrow transplantation (BMT) are largely unknown. To address this gap in knowledge, we used machine learning to decipher clinically relevant signatures from immunophenotypic, proteomic, and clinical data and then examined transcriptome changes in the lymphocyte subsets that predicted major posttransplant outcomes. Kinetics of immune subset reconstitution after day 28 were similar for 70 patients undergoing haplo and 75 patients undergoing HLA-matched BMT. Machine learning based on 35 candidate factors (10 clinical, 18 cellular, and 7 proteomic) revealed that combined elevations in effector CD4+ conventional T cells (Tconv) and CXCL9 at day 28 predicted acute graft-versus-host disease (aGVHD). Furthermore, higher NK cell counts predicted improved overall survival (OS) due to a reduction in both nonrelapse mortality and relapse. Transcriptional and flow-cytometric analyses of recovering lymphocytes in patients with aGVHD identified preserved hallmarks of functional CD4+ regulatory T cells (Tregs) while highlighting a Tconv-driven inflammatory and metabolic axis distinct from that seen with conventional GVHD prophylaxis. Patients developing early relapse displayed a loss of inflammatory gene signatures in NK cells and a transcriptional exhaustion phenotype in CD8+ T cells. Using a multimodality approach, we highlight the utility of systems biology in BMT biomarker discovery and offer a novel understanding of how PTCy influences alloimmune responses. Our work charts future directions for novel therapeutic interventions after these increasingly used GVHD prophylaxis platforms. Specimens collected on NCT0079656226 and NCT0080927627 https://clinicaltrials.gov/.


Subject(s)
Bone Marrow Transplantation , Cyclophosphamide/therapeutic use , Graft vs Host Disease/diagnosis , Immunosuppressive Agents/therapeutic use , Adult , Bone Marrow Transplantation/adverse effects , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Humans , Immune Reconstitution , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Machine Learning , Male , Middle Aged , Proteomics , Transcriptome , Young Adult
2.
Blood ; 132(20): 2188-2200, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30181175

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is a major complication of allogeneic hematopoietic cell transplantation (allo-HCT) and remains an area of unmet clinical need with few treatment options available. Notch blockade prevents acute GVHD in multiple mouse models, but the impact of Notch signaling on cGVHD remains unknown. Using genetic and antibody-mediated strategies of Notch inhibition, we investigated the role of Notch signaling in complementary mouse cGVHD models that mimic several aspects of human cGVHD in search of candidate therapeutics. In the B10.D2→BALB/c model of sclerodermatous cGVHD, Delta-like ligand 4 (Dll4)-driven Notch signaling was essential for disease development. Antibody-mediated Dll4 inhibition conferred maximum benefits when pursued early in a preventative fashion, with anti-Dll1 enhancing early protection. Notch-deficient alloantigen-specific T cells showed no early defects in proliferation or helper polarization in vivo but subsequently exhibited markedly decreased cytokine secretion and enhanced accumulation of FoxP3+ regulatory T cells. In the B6→B10.BR major histocompatibility complex-mismatched model with multi-organ system cGVHD and prominent bronchiolitis obliterans (BO), but not skin manifestations, absence of Notch signaling in T cells provided long-lasting disease protection that was replicated by systemic targeting of Dll1, Dll4, or both Notch ligands, even during established disease. Notch inhibition decreased target organ damage and germinal center formation. Moreover, decreased BO-cGVHD was observed upon inactivation of Notch1 and/or Notch2 in T cells. Systemic targeting of Notch2 alone was safe and conferred therapeutic benefits. Altogether, Notch ligands and receptors regulate key pathogenic steps in cGVHD and emerge as novel druggable targets to prevent or treat different forms of cGVHD.


Subject(s)
Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Intercellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , Receptors, Notch/immunology , Adaptor Proteins, Signal Transducing , Animals , Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/pathology , Calcium-Binding Proteins , Chronic Disease , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Isoantigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transplantation, Homologous/adverse effects
3.
FASEB J ; 33(3): 3330-3342, 2019 03.
Article in English | MEDLINE | ID: mdl-30383451

ABSTRACT

Rheumatoid arthritis (RA) is an inflammatory joint disease that eventually leads to permanent bone and cartilage destruction. Fas has already been established as the regulator of inflammation in RA, but its role in bone formation under arthritic conditions is not completely defined. The aim of this study was to assess the effect of Fas inactivation on the bone damage during murine antigen-induced arthritis. Subchondral bone of wild-type (WT) and Fas-knockout (Fas-/-) mice was evaluated by histomorphometry and microcomputerized tomography. Proportions of synovial bone and cartilage progenitors were assessed by flow cytometry. Synovial bone and cartilage progenitors were purified by fluorescence-activated cell sorting and expression of Fas and Fas-induced apoptosis were analyzed in vitro. Results showed that Fas-/- mice developed attenuated arthritis characterized by preserved epiphyseal bone and cartilage. A proportion of the earliest CD200+ bone and cartilage progenitors was reduced in WT mice with arthritis and was unaltered in Fas-/- mice. During osteoblastic differentiation in vitro, CD200+ cells express the highest levels of Fas and are removed by Fas ligation. These results suggest that Fas-induced apoptosis of early CD200+ osteoprogenitor population represents potential mechanism underlying the impaired bone formation in arthritis, so their preservation may represent the bone-protective mechanism during arthritis.-Lazic Mosler, E., Lukac, N., Flegar, D., Fadljevic, M., Radanovic, I., Cvija, H., Kelava, T., Ivcevic, S., Sucur, A., Markotic, A., Katavic, V., Marusic, A., Grcevic, D., Kovacic, N. Fas receptor induces apoptosis of synovial bone and cartilage progenitor populations and promotes bone loss in antigen-induced arthritis.


Subject(s)
Antigens/metabolism , Apoptosis/physiology , Arthritis, Rheumatoid/metabolism , Bone and Bones/metabolism , Cartilage/metabolism , Stem Cells/metabolism , Synovial Membrane/metabolism , fas Receptor/metabolism , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone and Bones/physiology , Cartilage/physiology , Cells, Cultured , Female , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Synovial Membrane/pathology
5.
Neuroimmunomodulation ; 21(5): 226-33, 2014.
Article in English | MEDLINE | ID: mdl-24603633

ABSTRACT

OBJECTIVES: Receptor for advanced glycation end products (RAGE) ligands/RAGE interactions have been proposed to have a pathogenic role in neuroinflammatory disorders. Our study aimed to assess changes in high-mobility group box (HMGB)1 and its receptor RAGE in peripheral blood (PBL) and cerebrospinal fluid (CSF) of patients with multiple sclerosis (MS) at the disease onset compared with control subjects. METHODS: PBL and CSF were collected from control subjects (n = 30) and MS patients (n = 27) at clinical onset. Soluble RAGE (sRAGE), HMGB1, S100 calcium-binding protein A12 (S100A12), interleukin (IL)-1ß and tumor necrosis factor (TNF)-α were measured in the CSF and plasma by enzyme-linked immunosorbent assay. Gene expression in PBL mononuclear cells (PBMCs) was detected by quantitative PCR for RAGE, HMGB1, S100A12 and several proinflammatory/immunoregulatory cytokines. RESULTS: We found a significantly lower expression of IL-10 (p = 0.031) in the PBMCs of MS patients. The level of sRAGE in the CSF of MS patients was lower (p = 0.021), with the ability to discriminate between MS patients and control subjects. Moreover, PBMC gene expression for HMGB1 and S100A12 positively correlated with IL-6. CONCLUSIONS: Our study confirmed that the cytokine network is disturbed in PBL and CSF at MS clinical onset. The deregulated HMGB1/RAGE axis found in our study may present an early pathogenic event in MS, proposing sRAGE as a possible novel therapeutic strategy for MS treatment.


Subject(s)
Biomarkers/analysis , HMGB1 Protein/blood , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Adult , Chemotactic Factors/blood , Chemotactic Factors/cerebrospinal fluid , Enzyme-Linked Immunosorbent Assay , Female , HMGB1 Protein/cerebrospinal fluid , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Receptor for Advanced Glycation End Products/blood , S100 Proteins/blood , S100 Proteins/cerebrospinal fluid , Transcriptome
6.
Int Orthop ; 38(1): 183-92, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24100919

ABSTRACT

PURPOSE: We aimed to assess osteoclastogenic potential of peripheral blood mononuclear cells (PBMC) and synovial fluid-derived mononuclear cells (SFMC) in different forms of arthritis and to correlate it with inflammatory mediators within intra-articular and circulatory compartments. METHODS: Paired PBMC and SFMC samples of patients with rheumatoid arthritis (RA; n = 10) and psoriatic arthritis (PsA; n = 10), and PBMC of healthy controls were cultured to assess osteoclastogenic potential by the number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts (OCs) and expression of OC-related genes (receptor activator of nuclear factor-κΒ (RANK), cFMS, and TRAP). Osteoclastogenesis was correlated with the arthritis-related inflammatory indicators in serum and synovial fluid (SF). RESULTS: Number of OCs differentiated from PBMC was significantly higher in RA and PsA compared with control, with RA having more OCs compared with PsA. There was no difference in SFMC OC number between arthritic patients, but RANK expression in OCs differentiated from SFMC was higher in PsA compared with RA. SF of PsA patients more potently induced OC differentiation from control CD3(-)CD19(-)CD56(-)CD11b(+)CD115(+) PBMC compared with RA, paralleled with higher RANK-ligand expression in PsA SFMC. Positive correlations of OC number with erythrocyte sedimentation rate, serum level of CCL2, and PBMC gene expression of interleukin-18 and Fas-ligand were observed. CONCLUSION: Osteoclastogenic potential is systemically enhanced in patients with RA, paralleled by disordered systemic and local expression of proinflammatory mediators, whereas PsA involves specific deregulation in RANKL/RANK axis. Our study reveals arthritis-specific mediators associated with the form of arthritis, indicating clinical relevance for diagnosis and treatment.


Subject(s)
Arthritis, Psoriatic/physiopathology , Arthritis, Rheumatoid/physiopathology , Cell Differentiation , Inflammation/metabolism , Leukocytes, Mononuclear/pathology , Osteoclasts/pathology , Severity of Illness Index , Synovial Fluid/cytology , Acid Phosphatase/metabolism , Adult , Aged , Arthritis, Psoriatic/metabolism , Arthritis, Psoriatic/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Case-Control Studies , Cell Count , Cells, Cultured , Female , Humans , Isoenzymes/metabolism , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Osteoclasts/metabolism , Predictive Value of Tests , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sensitivity and Specificity , Synovial Fluid/metabolism , Tartrate-Resistant Acid Phosphatase
7.
Connect Tissue Res ; 54(4-5): 296-304, 2013.
Article in English | MEDLINE | ID: mdl-23782451

ABSTRACT

Previous studies reported that embryonic stem cells (ESCs) can be induced to differentiate into cells showing a mature osteoblastic phenotype by culturing them under osteo-inductive conditions. It is probable that osteogenic differentiation requires that ESCs undergo differentiation through an intermediary step involving a mesenchymal lineage precursor. Based on our previous studies indicating that adult mesenchymal progenitor cells express α-smooth muscle actin (αSMA), we have generated ESCs from transgenic mice in which an αSMA promoter directs the expression of red fluorescent protein (RFP) to mesenchymal progenitor cells. To track the transition of ESC-derived MSCs into mature osteoblasts, we have utilized a bone-specific fragment of rat type I collagen promoter driving green fluorescent protein (Col2.3GFP). Following osteogenic induction in ESCs, we have observed expression of alkaline phosphatase (ALP) and subsequent mineralization as detected by von Kossa staining. After 1 week of osteogenic induction, ESCs begin to express αSMARFP. This expression was localized to the peripheral area encircling a typical ESC colony. Nevertheless, these αSMARFP positive cells did not show activation of the Col2.3GFP promoter, even after 7 weeks of osteogenic differentiation in vitro. In contrast, Col2.3GFP expression was detected in vivo, in mineralized areas following teratoma formation. Our results indicate that detection of ALP activity and mineralization of ESCs cultured under osteogenic conditions is not sufficient to demonstrate osteogenic maturation. Our study indicates the utility of the promoter-visual transgene approach to assess the commitment and differentiation of ESCs into the osteoblast lineage.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Osteogenesis/physiology , Alkaline Phosphatase/metabolism , Animals , Mice , Mice, Transgenic
8.
J Immunol ; 184(2): 764-74, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19995899

ABSTRACT

Donor CD4+ T cells are thought to be essential for inducing delayed host tissue injury in chronic graft-versus-host disease (GVHD). However, the relative contributions of distinct effector CD4+ T cell subpopulations and the molecular pathways influencing their generation are not known. We investigated the role of the STAT3 pathway in a murine model of chronic sclerodermatous GVHD. This pathway integrates multiple signaling events during the differentiation of naive CD4+ T cells and impacts their homeostasis. We report that chimeras receiving an allograft containing STAT3-ablated donor CD4+ T cells do not develop classic clinical and pathological manifestations of alloimmune tissue injury. Analysis of chimeras showed that abrogation of STAT3 signaling reduced the in vivo expansion of donor-derived CD4+ T cells and their accumulation in GVHD target tissues without abolishing antihost alloreactivity. STAT3 ablation did not significantly affect Th1 differentiation while enhancing CD4+CD25+Foxp3+ T cell reconstitution through thymus-dependent and -independent pathways. Transient depletion of CD25+ T cells in chimeras receiving STAT3-deficient T cells resulted in delayed development of alloimmune gut and liver injury. This delayed de novo GVHD was associated with the emergence of donor hematopoietic stem cell-derived Th1 and Th17 cells. These results suggest that STAT3 signaling in graft CD4+ T cells links the alloimmune tissue injury of donor graft T cells and the emergence of donor hematopoietic stem cell-derived pathogenic effector cells and that both populations contribute, albeit in different ways, to the genesis of chronic GVHD after allogenic bone marrow transplantation in a murine model.


Subject(s)
Bone Marrow Transplantation/immunology , CD4-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/etiology , STAT3 Transcription Factor/metabolism , Scleroderma, Systemic/etiology , Animals , Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes/immunology , Chronic Disease , Disease Models, Animal , Mice , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Transplantation, Homologous
9.
Clin Epigenetics ; 9: 106, 2017.
Article in English | MEDLINE | ID: mdl-29026447

ABSTRACT

Aortic valve stenosis is the most common cardiac valve disease, and with current trends in the population demographics, its prevalence is likely to rise, thus posing a major health and economic burden facing the worldwide societies. Over the past decade, it has become more than clear that our traditional genetic views do not sufficiently explain the well-known link between AS, proatherogenic risk factors, flow-induced mechanical forces, and disease-prone environmental influences. Recent breakthroughs in the field of epigenetics offer us a new perspective on gene regulation, which has broadened our perspective on etiology of aortic stenosis and other aortic valve diseases. Since all known epigenetic marks are potentially reversible this perspective is especially exciting given the potential for development of successful and non-invasive therapeutic intervention and reprogramming of cells at the epigenetic level even in the early stages of disease progression. This review will examine the known relationships between four major epigenetic mechanisms: DNA methylation, posttranslational histone modification, ATP-dependent chromatin remodeling, and non-coding regulatory RNAs, and initiation and progression of AS. Numerous profiling and functional studies indicate that they could contribute to endothelial dysfunctions, disease-prone activation of monocyte-macrophage and circulatory osteoprogenitor cells and activation and osteogenic transdifferentiation of aortic valve interstitial cells, thus leading to valvular inflammation, fibrosis, and calcification, and to pressure overload-induced maladaptive myocardial remodeling and left ventricular hypertrophy. This is especcialy the case for small non-coding microRNAs but was also, although in a smaller scale, convincingly demonstrated for other members of cellular epigenome landscape. Equally important, and clinically most relevant, the reported data indicate that epigenetic marks, particularly certain microRNA signatures, could represent useful non-invasive biomarkers that reflect the disease progression and patients prognosis for recovery after the valve replacement surgery.


Subject(s)
Aortic Valve Stenosis/genetics , Epigenesis, Genetic , Hypertrophy, Left Ventricular/genetics , MicroRNAs/genetics , Chromatin Assembly and Disassembly , DNA Methylation , Disease Progression , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Protein Processing, Post-Translational
10.
Arthritis Res Ther ; 19(1): 142, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28619088

ABSTRACT

BACKGROUND: The peripheral blood (PB) monocyte pool contains osteoclast progenitors (OCPs), which contribute to osteoresorption in inflammatory arthritides and are influenced by the cytokine and chemokine milieu. We aimed to define the importance of chemokine signals for migration and activation of OCPs in rheumatoid arthritis (RA) and psoriatic arthritis (PsA). METHODS: PB and, when applicable, synovial fluid (SF) samples were collected from 129 patients with RA, 53 patients with PsA, and 110 control patients in parallel to clinical parameters of disease activity, autoantibody levels, and applied therapy. Receptors for osteoclastogenic factors (CD115 and receptor activator of nuclear factor-κB [RANK]) and selected chemokines (CC chemokine receptor 1 [CCR1], CCR2, CCR4, CXC chemokine receptor 3 [CXCR3], CXCR4) were determined in an OCP-rich subpopulation (CD3-CD19-CD56-CD11b+CD14+) by flow cytometry. In parallel, levels of CC chemokine ligand 2 (CCL2), CCL3, CCL4, CCL5, CXC chemokine ligand 9 (CXCL9), CXCL10, and CXCL12 were measured using cytometric bead array or enzyme-linked immunosorbent assay. Sorted OCPs were stimulated in culture by macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand, and they were differentiated into mature osteoclasts that resorb bone. Selected chemokines (CCL2, CCL5, CXCL10, and CXCL12) were tested for their osteoclastogenic and chemotactic effects on circulatory OCPs in vitro. RESULTS: The OCP population was moderately enlarged among PB cells in RA and correlated with levels of tumor necrosis factor-α (TNF-α), rheumatoid factor, CCL2, and CCL5. Compared with PB, the RANK+ subpopulation was expanded in SF and correlated with the number of tender joints. Patients with PsA could be distinguished by increased RANK expression rather than total OCP population. OCPs from patients with arthritis had higher expression of CCR1, CCR2, CCR4, CXCR3, and CXCR4. In parallel, patients with RA had increased levels of CCL2, CCL3, CCL4, CCL5, CXCL9, and CXCL10, with significant elevation in SF vs PB for CXCL10. The subset expressing CXCR4 positively correlated with TNF-α, bone resorption marker, and rheumatoid factor, and it was reduced in patients treated with disease-modifying antirheumatic drugs. The CCR4+ subset showed a significant negative trend during anti-TNF treatment. CCL2, CCL5, and CXCL10 had similar osteoclastogenic effects, with CCL5 showing the greatest chemotactic action on OCPs. CONCLUSIONS: In our study, we identified distinct effects of selected chemokines on stimulation of OCP mobilization, tissue homing, and maturation. Novel insights into migratory behaviors and functional properties of circulatory OCPs in response to chemotactic signals could open ways to new therapeutic targets in RA.


Subject(s)
Arthritis, Psoriatic/pathology , Arthritis, Rheumatoid/pathology , Cell Differentiation , Chemokines/metabolism , Osteoclasts/pathology , Stem Cells/pathology , Adult , Arthritis, Psoriatic/immunology , Arthritis, Psoriatic/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/physiology , Cell Movement/physiology , Chemokines/immunology , Female , Humans , Male , Middle Aged , Osteoclasts/metabolism , Stem Cells/metabolism
11.
Exp Hematol ; 42(11): 966-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25051532

ABSTRACT

Osteoclasts are components of hematopoietic stem cell (HSC) niches, but their role as contributors to the HSC homeostasis and release are still controversial. We aimed to investigate whether an acute blood loss of 10% of total blood content, along with the consequent intense hematopoiesis, would affect osteoclast differentiation and activity. Isolated peripheral blood, spleen, and bone marrow (BM) cells from bones of hind limbs were investigated for the presence of specific subpopulations of osteoclast precursors: B220(-)CD3(-)NK1.1(-)CD11b(-/low)CD115(+)CD117(+) cells in BM, and B220(-)CD3(-)NK1.1(-)Gr-1(-)CD11b(+)CD115(+) cells in peripheral blood and spleen as well as the receptor activator of nuclear factor κ-B(+) cycle-arrested quiescent osteoclast precursors. Expression of osteoclastogenesis-related genes CD115, receptor activator of nuclear factor κ-B, and cathepsin K, the potential of BM cells to form osteoclast-like cells in vitro, and osteoclast activity in vivo were also evaluated. We observed an increase in spleen cellularity and myelopoiesis during week 1 following blood loss, without any significant effects on BM cellularity or BM myeloid precursors, including cells with high osteoclastogenic potential. However, at 1 week postbleeding, hematopoiesis significantly promoted the expression of cathepsin K, interleukin-34, and bone morphogenetic protein-6. Quiescent osteoclast precursors increased significantly in spleen 2 days following bleeding, whereas osteoclast activity remained unchanged up to 2 weeks postbleeding. Osteoclast-dependent B-cell differentiation was affected at the pre-B stage of maturation in BM, whereas the Lin(-)Sca-1(+)c-kit(+) population expanded in BM and spleen after 2 days postbleeding. Our data demonstrate that an acute blood loss promotes differentiation and maturation of osteoclasts at 1 week but does not enhance osteoresorption at 2 weeks postbleeding. Our data also identify osteoclast differentiation as a consequent and important event in establishing HSC homeostasis following hematopoietic stress.


Subject(s)
Bone Marrow/metabolism , Hematopoiesis/genetics , Osteoclasts/cytology , Stress, Physiological , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Ly/genetics , Antigens, Ly/metabolism , Biomarkers/metabolism , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Cathepsin K/genetics , Cathepsin K/metabolism , Cell Differentiation , Cell Proliferation , Female , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukins/genetics , Interleukins/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , Phlebotomy , Primary Cell Culture , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Spleen/cytology , Spleen/metabolism
12.
Leuk Res ; 37(6): 705-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23528261

ABSTRACT

We investigated the role of bone morphogenetic proteins (BMPs) in suppression of all-trans retinoic acid (ATRA)-mediated differentiation of leukemic promyelocytes. In NB4 and HL60 cell lines, BMPs reduced the percentage of differentiated cells, and suppressed PU.1 and C/EBPε gene expression induced by ATRA. BMP and ATRA synergized in the induction of ID genes, causing suppression of differentiation. In primary acute promyelocytic leukemia bone-marrow samples, positive correlation of PML/RARα and negative of RARα with the expression of BMP-4, BMP-6 and ID genes were found. We concluded that BMPs may have oncogenic properties and mediate ATRA resistance by a mechanism that involves ID genes.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/drug effects , Granulocyte Precursor Cells/drug effects , Leukemia, Promyelocytic, Acute/physiopathology , Neoplastic Stem Cells/drug effects , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/physiology , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 4/physiology , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein 6/physiology , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/genetics , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/physiology , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured
13.
Int J Cardiol ; 164(1): 7-20, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-22243936

ABSTRACT

Cardiac myxomas are rare benign and slowly proliferating neoplasms of uncertain histogenesis with heterogeneous histomorphology and variable and sometimes clinically quite malignant pathological manifestations. Majority of cardiac myxoma occur sporadically while a relatively small proportion of diagnosed cases develop as a part of Carney complex syndrome with established familial pattern of inheritance. Although histologically indistinguishable these two forms of cardiac myxoma exhibit distinct cytogenetic make-up and apparent pathological differences important for their clinical presentation and prognosis. Additional problem is presented with secondary lesions with more aggressive histology and significantly faster cell proliferation suggesting their successive malignant alteration. Surgical resection of cardiac myxoma is currently the only treatment of choice. However, to avoid potentially hazardous operating procedures and possible postoperative complications and to prevent recurrence of the neoplastic lesions it is necessary to develop alternative approaches and identify a possible drug targets for their successful pharmacological treatment. Due to the rarity of the disease, a small number of cases in one institution and lack of comprehensive experimental data particularly concerning the cases of metastatic dissemination and secondary lesions with malignant nature, a comprehensive multi-institutional approach is required for better understanding of their molecular pathology and illumination of key molecular, genetic as well as epigenetic markers and regulatory pathways responsible for their development. In this article we provide comprehensive pathohistological, molecular and cytogenetic overview of sporadic cardiac myxoma cases restating the major hypothesis concerning their histogenesis and emphasizing potential approaches for their further reexamination.


Subject(s)
Heart Neoplasms/pathology , Molecular Diagnostic Techniques , Myxoma/pathology , Diagnosis, Differential , Heart Neoplasms/diagnosis , Heart Neoplasms/etiology , Humans , Microarray Analysis , Myxoma/diagnosis , Myxoma/etiology , Neoplasm Recurrence, Local
14.
Inflammation ; 35(5): 1618-31, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22699680

ABSTRACT

In our study, we explored the bidirectional communication via soluble factors between bone cells and endotoxin-stimulated splenic lymphocytes in an in vitro coculture model that mimics the inflammatory environment. Both the ability of lymphocytes to affect differentiation and immune properties of bone cells, osteoblasts (OBL) and osteoclasts (OCL), and of bone cells to modulate cytokine and activation profile of endotoxin-stimulated lymphocytes were tested. LPS-pulsed lymphocytes enhanced OCL but inhibited OBL differentiation and increased the RANKL/OPG ratio, and, at the same time, upregulated chemotactic properties of bone cells, specifically CCL2, CCL5, and CXCL10 in OCL and CCL5 and CXCL13 in OBL. In parallel, bone cells had immunosuppressive effects by downregulating the lymphocyte expression of interleukin (IL)-1, IL-6, TNF-α and co-stimulatory molecules. OCL stimulated the production of osteoclastogenic cytokine RANKL in T lymphocytes. The anti-inflammatory effect, especially of OBL, suggests a possible compensatory mechanism to limit the inflammatory reaction during infection.


Subject(s)
Lymphocyte Activation , Lymphocytes/immunology , Osteoblasts/immunology , Osteoclasts/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemokine CCL5/biosynthesis , Chemokine CXCL10 , Chemokine CXCL13/biosynthesis , Female , Inflammation , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoprotegerin/biosynthesis , RANK Ligand/biosynthesis , RANK Ligand/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
15.
J Rheumatol ; 37(2): 246-56, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20008919

ABSTRACT

OBJECTIVE: To assess whether different forms of arthritis and disease activity could be distinguished by peripheral blood expression profiles of bone-regulatory factors including tumor necrosis factor (TNF)-superfamily [TNF-related apoptosis-inducing ligand (TRAIL), the Fas ligand (FasL), and the ligand for herpesvirus entry mediator (LIGHT)] and bone morphogenetic protein (BMP)-family members (BMP-2, BMP-4, BMP-6) as well as osteoblast differentiation gene Runx2. METHODS: Blood cells from healthy controls (n = 25) and patients at different disease stages with rheumatoid arthritis (RA; n = 49), osteoarthritis (OA; n = 17), or spondyloarthritis, including ankylosing spondylitis (AS; n = 27) or psoriatic arthritis (PsA; n = 23), were processed for quantitative polymerase chain reaction. Gene expression was assessed in comparison with control samples, correlated with clinical data of different forms of arthritis, and analyzed for discriminative efficacy between groups by receiver-operation characteristic (ROC) curves. Results were confirmed on diagnostic RA (n = 5) and AS (n = 8) samples. RESULTS: BMP-4, BMP-6, and Runx2 expressions were significantly decreased in patients with RA and OA versus controls. Patients with RA also had decreased FasL and LIGHT expression, while patients with AS had increased Runx2 expression. Negative correlation with disease activity was found for BMP-4, FasL, and Runx2 in RA and for Runx2 in PsA, while positive correlation was found for BMP-4 in PsA. Gene expression was higher in the therapy-resistant form of AS (for BMP-4, LIGHT, and Runx2) and in methotrexate-treated patients in RA (for BMP-2 and LIGHT). ROC curve analysis confirmed discrimination between groups, particularly decreased LIGHT and Runx2 for RA and increased Runx2 for AS. CONCLUSION: Our study identified BMP and Runx2 as possible biomarkers of bone metabolism in several forms of arthritis, while lower FasL and LIGHT were associated with RA. Correlation between gene expression and disease activity may be clinically useful in assessing therapeutic effectiveness and disease monitoring.


Subject(s)
Arthritis/diagnosis , Bone Morphogenetic Proteins/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Severity of Illness Index , Tumor Necrosis Factors/genetics , Adult , Aged , Analysis of Variance , Arthritis/genetics , Arthritis/immunology , Biomarkers/analysis , Bone Morphogenetic Proteins/immunology , Core Binding Factor Alpha 1 Subunit/immunology , Female , Gene Expression , Humans , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/immunology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/immunology , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Tumor Necrosis Factors/immunology
16.
Leuk Res ; 34(6): 742-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19926132

ABSTRACT

We assessed the expression pattern and clinical relevance of BMPs and related molecules in multiple myeloma (MM). MM bone-marrow samples (n=32) had increased BMP4, BMP6, ACVR1 and ACVR2A, and decreased NOG expression compared with controls (n=15), with BMP6 having the highest sensitivity/specificity. Within MM bone-marrow, the source of BMPs was mainly CD138(+) plasma-cell population, and BMP6 and ACVR1 expression correlated with plasma-cell percentage. Using myeloma cell lines NCI H929 and Thiel we showed that BMPs induced ID1, ID2 and IL6, and suppressed CDKN1A and BAX gene expression, and BAX protein expression. Finally, BMPs partially protected myeloma cells from bortezomib- and TRAIL-induced apoptosis. We concluded that BMPs may be involved in MM pathophysiology and serve as myeloma cell biomarkers.


Subject(s)
Bone Marrow Cells/metabolism , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Proteins/genetics , Inhibitor of Differentiation Proteins/genetics , Multiple Myeloma/genetics , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/pathology , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , Case-Control Studies , Cell Proliferation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Proteins/metabolism , Male , Middle Aged , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured , Up-Regulation/genetics
17.
Cell Immunol ; 233(2): 133-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15950207

ABSTRACT

We have previously shown that mice with generalised lymphoproliferative disorder (gld) have increased bone mass in addition to autoimmune disease characterised by the accumulation of double negative (dn) T lymphocytes (CD3(+)CD4(-)CD8(-)CD45R(+)). To further explore the association of the immune disorder with the bone phenotype of gld mice, we established parabiotic circulation between gld and wild-type animals (C57BL/6, B6). One week after the surgery, the proportion of dn T lymphocytes increased in peripheral blood, bone marrow, spleen, and lymph nodes of wild-type members of the B6-gld parabiotic pair and decreased in tissues of gld pair members. The mixing of cells continued during four weeks of parabiosis. Number of osteoclast-like (OCL) cells in bone marrow cultures from a wild-type member of B6-gld parabiotic pair at the end of the first week decreased from 266+/-52 to 120+/-5OCL/cm(2), P<0.05, comparable with gld mice (99+/-21OCL/cm(2)), while the number of osteoblast colonies did not change. After four weeks, number of OCL cells formed from the bone marrow of B6 parabiotic mice was still similar to the number of OCL cells in their gld counterparts (150+/-18 and 131+/-24OCL/cm(2), respectively). In addition, the number of osteoblast colonies in B6 members of B6-gld parabiotic pairs increased (from 6+/-2 to 18+/-1colonies/cm(2), P<0.05) thus resembling the cell cultures of gld mice (18+/-1colonies/cm(2)). Taken together, these data show that the circulation of cells, including dn T lymphocytes established by parabiosis confers the osteoclast and osteoblast phenotype of gld to wild-type animals.


Subject(s)
Bone and Bones/blood supply , Bone and Bones/metabolism , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Parabiosis , Phenotype , Animals , Blood Circulation/genetics , Blood Circulation/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone and Bones/immunology , Cells, Cultured , Disease Models, Animal , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/physiopathology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL