Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Rheumatology (Oxford) ; 62(10): 3469-3479, 2023 10 03.
Article in English | MEDLINE | ID: mdl-36802235

ABSTRACT

OBJECTIVE: Trained immunity (TI) is a de facto memory program of innate immune cells, characterized by immunometabolic and epigenetic changes sustaining enhanced production of cytokines. TI evolved as a protective mechanism against infections; however, inappropriate activation can cause detrimental inflammation and might be implicated in the pathogenesis of chronic inflammatory diseases. In this study, we investigated the role of TI in the pathogenesis of giant cell arteritis (GCA), a large-vessel vasculitis characterized by aberrant macrophage activation and excess cytokine production. METHODS: Monocytes from GCA patients and from age- and sex-matched healthy donors were subjected to polyfunctional studies, including cytokine production assays at baseline and following stimulation, intracellular metabolomics, chromatin immunoprecipitation-qPCR, and combined ATAC/RNA sequencing. Immunometabolic activation (i.e. glycolysis) was assessed in inflamed vessels of GCA patients with FDG-PET and immunohistochemistry (IHC), and the role of this pathway in sustaining cytokine production was confirmed with selective pharmacologic inhibition in GCA monocytes. RESULTS: GCA monocytes exhibited hallmark molecular features of TI. Specifically, these included enhanced IL-6 production upon stimulation, typical immunometabolic changes (e.g. increased glycolysis and glutaminolysis) and epigenetic changes promoting enhanced transcription of genes governing pro-inflammatory activation. Immunometabolic changes of TI (i.e. glycolysis) were a feature of myelomonocytic cells in GCA lesions and were required for enhanced cytokine production. CONCLUSIONS: Myelomonocytic cells in GCA activate TI programs sustaining enhanced inflammatory activation with excess cytokine production.


Subject(s)
Giant Cell Arteritis , Humans , Giant Cell Arteritis/pathology , Monocytes/metabolism , Trained Immunity , Inflammation , Cytokines
2.
STAR Protoc ; 3(3): 101601, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35990738

ABSTRACT

In this protocol, we describe how to generate 3D culture surrogates of chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) bone marrow microenvironments. We detail the use of culturing scaffolds populated with BM stromal cells and tumor cells in the RCCS™ bioreactor. This 3D culture can efficiently recapitulate tumor-stroma crosstalk and allows the testing of drugs such as ibrutinib and bortezomib. Moreover, this protocol can be used for the generation of other and more complex tumor microenvironments. For complete details on the use and execution of this protocol, please refer to Belloni et al. (2018) and Barbaglio et al. (2021).


Subject(s)
Bone Marrow , Multiple Myeloma , Bone Marrow/pathology , Bortezomib , Cell Culture Techniques , Humans , Multiple Myeloma/pathology , Stromal Cells/pathology , Tumor Microenvironment
3.
Antibodies (Basel) ; 11(3)2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35892706

ABSTRACT

Therapeutic monoclonal antibodies (mAbs) are an emerging and very active frontier in clinical oncology, with hundred molecules currently in use or being tested. These treatments have already revolutionized clinical outcomes in both solid and hematological malignancies. However, identifying patients who are most likely to benefit from mAbs treatment is currently challenging and limiting the impact of such therapies. To overcome this issue, and to fulfill the expectations of mAbs therapies, it is urgently required to develop proper culture models capable of faithfully reproducing the interactions between tumor and its surrounding native microenvironment (TME). Three-dimensional (3D) models which allow the assessment of the impact of drugs on tumors within its TME in a patient-specific context are promising avenues to progressively fill the gap between conventional 2D cultures and animal models, substantially contributing to the achievement of personalized medicine. This review aims to give a brief overview of the currently available 3D models, together with their specific exploitation for therapeutic mAbs testing, underlying advantages and current limitations to a broader use in preclinical oncology.

5.
Blood ; 138(17): 1554-1569, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34077954

ABSTRACT

Trained immunity (TI) is a proinflammatory program induced in monocyte/macrophages upon sensing of specific pathogens and is characterized by immunometabolic and epigenetic changes that enhance cytokine production. Maladaptive activation of TI (ie, in the absence of infection) may result in detrimental inflammation and development of disease; however, the exact role and extent of inappropriate activation of TI in the pathogenesis of human diseases is undetermined. In this study, we uncovered the oncogene-induced, maladaptive induction of TI in the pathogenesis of a human inflammatory myeloid neoplasm (Erdheim-Chester disease, [ECD]), characterized by the BRAFV600E oncogenic mutation in monocyte/macrophages and excess cytokine production. Mechanistically, myeloid cells expressing BRAFV600E exhibit all molecular features of TI: activation of the AKT/mammalian target of rapamycin signaling axis; increased glycolysis, glutaminolysis, and cholesterol synthesis; epigenetic changes on promoters of genes encoding cytokines; and enhanced cytokine production leading to hyperinflammatory responses. In patients with ECD, effective therapeutic strategies combat this maladaptive TI phenotype; in addition, pharmacologic inhibition of immunometabolic changes underlying TI (ie, glycolysis) effectively dampens cytokine production by myeloid cells. This study revealed the deleterious potential of inappropriate activation of TI in the pathogenesis of human inflammatory myeloid neoplasms and the opportunity for inhibition of TI in conditions characterized by maladaptive myeloid-driven inflammation.


Subject(s)
Erdheim-Chester Disease/genetics , Inflammation/genetics , Proto-Oncogene Proteins B-raf/genetics , Cells, Cultured , Epigenesis, Genetic , Erdheim-Chester Disease/immunology , Erdheim-Chester Disease/pathology , Humans , Immunity , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Oncogenes , Point Mutation , Proto-Oncogene Proteins B-raf/immunology
7.
Haematologica ; 105(10): 2440-2447, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33054085

ABSTRACT

Therapeutic strategies designed to tinker with cancer cell DNA damage response have led to the widespread use of PARP inhibitors for BRCA1/2-mutated cancers. In the haematological cancer multiple myeloma, we sought to identify analogous synthetic lethality mechanisms that could be leveraged upon established cancer treatments. The combination of ATR inhibition using the compound VX-970 with a drug eliciting interstrand cross-links, melphalan, was tested in in vitro, ex vivo, and most notably in vivo models. Cell proliferation, induction of apoptosis, tumor growth and animal survival were assessed. The combination of ATM inhibition with a drug triggering double strand breaks, doxorucibin, was also probed. We found that ATR inhibition is strongly synergistic with melphalan, even in resistant cells. The combination was dramatically effective in targeting myeloma primary patient cells and cell lines reducing cell proliferation and inducing apoptosis. The combination therapy significantly reduced tumor burden and prolonged survival in animal models. Conversely, ATM inhibition only marginally impacted on myeloma cell survival, even in combination with doxorucibin at high doses. These results indicate that myeloma cells extensively rely on ATR, but not on ATM, for DNA repair. Our findings posit that adding an ATR inhibitor such as VX-970 to established therapeutic regimens may provide a remarkably broad benefit to myeloma patients.


Subject(s)
Multiple Myeloma , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cell Survival , DNA Damage , DNA Repair , Humans , Melphalan/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics
8.
Cell Commun Signal ; 18(1): 156, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32967672

ABSTRACT

BACKGROUND: Targeted therapy with BRAF and MEK inhibitors has improved the survival of patients with BRAF-mutated metastatic melanoma, but most patients relapse upon the onset of drug resistance induced by mechanisms including genetic and epigenetic events. Among the epigenetic alterations, microRNA perturbation is associated with the development of kinase inhibitor resistance. Here, we identified and studied the role of miR-146a-5p dysregulation in melanoma drug resistance. METHODS: The miR-146a-5p-regulated NFkB signaling network was identified in drug-resistant cell lines and melanoma tumor samples by expression profiling and knock-in and knock-out studies. A bioinformatic data analysis identified COX2 as a central gene regulated by miR-146a-5p and NFkB. The effects of miR-146a-5p/COX2 manipulation were studied in vitro in cell lines and with 3D cultures of treatment-resistant tumor explants from patients progressing during therapy. RESULTS: miR-146a-5p expression was inversely correlated with drug sensitivity and COX2 expression and was reduced in BRAF and MEK inhibitor-resistant melanoma cells and tissues. Forced miR-146a-5p expression reduced COX2 activity and significantly increased drug sensitivity by hampering prosurvival NFkB signaling, leading to reduced proliferation and enhanced apoptosis. Similar effects were obtained by inhibiting COX2 by celecoxib, a clinically approved COX2 inhibitor. CONCLUSIONS: Deregulation of the miR-146a-5p/COX2 axis occurs in the development of melanoma resistance to targeted drugs in melanoma patients. This finding reveals novel targets for more effective combination treatment. Video Abstract.


Subject(s)
Cyclooxygenase 2/metabolism , Drug Resistance, Neoplasm , Inflammation Mediators/metabolism , Melanoma/drug therapy , Melanoma/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Protein Kinase Inhibitors/therapeutic use , Cell Line, Tumor , Cyclooxygenase 2/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/pathology , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
9.
J Leukoc Biol ; 108(2): 591-599, 2020 08.
Article in English | MEDLINE | ID: mdl-32056262

ABSTRACT

Erdheim-Chester disease (ECD) is a rare histiocytosis characterized by infiltration of multiple tissues by CD68+ foamy Mϕs (or 'histiocytes'). Clinical manifestations arise from mass-forming lesions or from tissue and systemic inflammation. ECD histiocytes harbor oncogenic mutations along the MAPK-kinase signaling pathway (BRAFV600E in more than half of the patients), and secrete abundant pro-inflammatory cytokines and chemokines. Based on these features, ECD is considered an inflammatory myeloid neoplasm, and is accordingly managed with targeted kinase inhibitors or immunosuppressive and cytokine-blocking agents. Evidence is emerging that maladaptive metabolic changes, particularly up-regulated glycolysis, represent an additional, mutation-driven feature of ECD histiocytes, which sustains deregulated and protracted pro-inflammatory activation and cytokine production. Besides translational relevance to the management of ECD patients and to the development of new therapeutic approaches, recognition of ECD as a natural human model of chronic, maladaptive Mϕ activation instructs the understanding of Mϕ dysfunction in other chronic inflammatory conditions.


Subject(s)
Disease Susceptibility , Erdheim-Chester Disease/etiology , Erdheim-Chester Disease/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Signal Transduction , Animals , Cellular Reprogramming , Energy Metabolism , Erdheim-Chester Disease/diagnosis , Erdheim-Chester Disease/therapy , Histiocytes/immunology , Histiocytes/metabolism , Histiocytes/pathology , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Macrophage Activation/genetics , Mutation , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Oncogenes
11.
Oncoimmunology ; 7(7): e1440929, 2018.
Article in English | MEDLINE | ID: mdl-29900045

ABSTRACT

Erdheim-Chester disease (ECD) is a rare histiocytosis, characterized by xanthogranulomatous tissue infiltration by foamy histiocytes. Fibrosis, a histologic hallmark of ECD, is responsible for lesion growth and clinical manifestations. Unraveling molecular fibrotic pathway in ECD would allow the identification of new pharmacologic targets. In this study, we evaluated serum and tissue samples from a large cohort of ECD patients focusing on two major pro-fibrotic mediators, TGF-ß1 and chemokine ligand 18 (CCL18). We found a marked increase in CCL18 but not TGF-ß1 levels in serum and lesions of ECD patients (p < 0.001), independently of treatment status and consistently over time. Using a linear mathematical model, we also found that elevated CCL18 serum levels correlate with both number and severity of disease localizations. These findings suggest the involvement of CCL18-induced fibrosis in ECD pathogenesis, providing a rationale for exploring CCL18 inhibition as a treatment for progressive fibrosis in ECD.

12.
Haematologica ; 103(4): 707-716, 2018 04.
Article in English | MEDLINE | ID: mdl-29326121

ABSTRACT

Multiple myeloma develops primarily inside the bone marrow microenvironment, that confers pro-survival signals and drug resistance. 3D cultures that reproduce multiple myeloma-bone marrow interactions are needed to fully investigate multiple myeloma pathogenesis and response to drugs. To this purpose, we exploited the 3D Rotary Cell Culture System bioreactor technology for myeloma-bone marrow co-cultures in gelatin scaffolds. The model was validated with myeloma cell lines that, as assessed by histochemical and electron-microscopic analyses, engaged contacts with stromal cells and endothelial cells. Consistently, pro-survival signaling and also cell adhesion-mediated drug resistance were significantly higher in 3D than in 2D parallel co-cultures. The contribution of the VLA-4/VCAM1 pathway to resistance to bortezomib was modeled by the use of VCAM1 transfectants. Soluble factor-mediated drug resistance could be also demonstrated in both 2D and 3D co-cultures. The system was then successfully applied to co-cultures of primary myeloma cells-primary myeloma bone marrow stromal cells from patients and endothelial cells, allowing the development of functional myeloma-stroma interactions and MM cell long-term survival. Significantly, genomic analysis performed in a high-risk myeloma patient demonstrated that culture in bioreactor paralleled the expansion of the clone that ultimately dominated in vivo Finally, the impact of bortezomib on myeloma cells and on specialized functions of the microenvironment could be evaluated. Our findings indicate that 3D dynamic culture of reconstructed human multiple myeloma microenvironments in bioreactor may represent a useful platform for drug testing and for studying tumor-stroma molecular interactions.


Subject(s)
Bone Marrow/pathology , Cell Communication , Cell Culture Techniques , Models, Biological , Multiple Myeloma/pathology , Bioreactors , Bortezomib/pharmacology , Cell Adhesion , Cell Survival , Coculture Techniques , Drug Resistance , Endothelial Cells , Gelatin , Humans , Multiple Myeloma/drug therapy , Stromal Cells , Tumor Microenvironment
13.
Oncoimmunology ; 6(6): e1318237, 2017.
Article in English | MEDLINE | ID: mdl-28680751

ABSTRACT

Treatment of Erdheim-Chester disease (ECD), a rare non-Langerhans histiocytosis, relies on interferon-α, chemotherapeutic agents such as purine analogs, cytokine-blocking agents and BRAF inhibitors. Since interleukin (IL)-6 levels are elevated in serum and lesions of ECD patients, we evaluated the therapeutic efficacy and safety of IL-6 blockade with tocilizumab. We conducted an open-label, single-arm, phase II, prospective study of tocilizumab in three patients with multisystem ECD and poor tolerance/contraindications to IFN-α. Modifications of symptoms attributed to ECD represented the criteria for evaluation of clinical response. Changes at positron emission tomography scan, computed tomography scan, and magnetic resonance imaging at month 6 represented the main criteria for the evaluation of radiological response. Sustained complete clinical response and partial radiological improvement were observed in two patients, paralleled by modulation of systemic pro-inflammatory mediators. In spite of disease stabilization or improvement at extra-neurological sites, a third patient experienced a radiologic and clinical progression of central nervous system involvement, mirrored by a dramatic increase of circulating IL-6 and related cytokines. These findings indicate that IL-6 inhibition can be effective in ECD, but caution is advisable in patients with neurologic involvement. IL-6 emerges as a central mediator in ECD pathogenesis.

14.
Methods Mol Biol ; 1612: 177-190, 2017.
Article in English | MEDLINE | ID: mdl-28634943

ABSTRACT

3D-dynamic culture models represent an invaluable tool for a better comprehension of tumor biology and drug response, as they accurately re-create/preserve the complex multicellular organization and the dynamic interactions of the parental microenvironment, which can affect tumor fate and drug sensitivity. Hence, development of models that recapitulate tumor within its embedding microenvironment is an imperative need. This is particularly true for multiple myeloma (MM), which survives almost exclusively in the bone marrow (BM). To meet this need, we have previously exploited and validated an innovative 3D-dynamic culture technology, based on the use of the Rotary Cell Culture System (RCCS ™) bioreactor . Here, we describe, step by step, the procedures we have employed to establish two human MM ex vivo models, i.e., the culture of human BM-derived isolated cells and of MM tissues from patients.


Subject(s)
Cell Culture Techniques/instrumentation , Models, Biological , Multiple Myeloma/pathology , Bone Marrow/pathology , Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Line, Tumor , Humans , Tissue Engineering , Tumor Microenvironment
15.
Oncoimmunology ; 5(7): e1181244, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27622037

ABSTRACT

Erdheim-Chester disease (ECD) is a rare non-Langerhans cell histiocytosis (LCH) characterized by tissue infiltration with CD68(+) foamy histiocytes. TNF-related chronic inflammation and mutations in the MAP kinase signaling pathway in histiocytes are recognized as the two major pathogenic events. Among pleomorphic clinical manifestations, cardiovascular involvement is frequent and prognostically relevant. Evaluation of ECD clinical course and response to treatment is, however, still challenging. Taking advantage of the two largest cohorts of ECD patients worldwide, we investigated the relevance and the potential of circulating Chromogranin A (CgA), a pro-hormone involved in cardiovascular homeostasis and inflammation, as a biomarker of response to therapy in ECD. Consistent with other TNF-related inflammatory diseases, we found that not only TNF-α and soluble TNF-Receptors (sTNF-Rs), but also CgA plasma levels were significantly increased in ECD patients compared to controls. CgA, but not sTNF-Rs, discriminated cardiovascular involvement in ECD patients and correlated with pro-Brain Natriuretic Peptide (pro-BNP). In a single case, where a cardiac biopsy was available, CgA was found expressed by cardiomyocytes but not by infiltrating histiocytes. In four ECD patients, where serial determination of these parameters was obtained, the kinetics of sTNF-Rs and CgA paralleled response to therapy with anti-cytokine inhibitors; specifically, sTNF-Rs overlapped TNF-associated inflammation, while CgA, together with pro-BNP, closely mirrored response of cardiac disease. Our data indicate that both sTNF-Rs and CgA are linked to ECD pathophysiology. Moreover, CgA, in concert with pro-BNP, can be further exploited to fulfill the unmet clinical need of non-invasive reliable biomarkers of cardiac disease in these patients.

16.
Cancer Res ; 76(7): 1781-91, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26869462

ABSTRACT

Angiogenesis has been postulated to be critical for the pathogenesis of multiple myeloma, a neoplastic disease characterized by abnormal proliferation of malignant plasma cells in the bone marrow (BM). Cleavage of the N- and C-terminal regions of circulating chromogranin A (CgA, CHGA), classically an antiangiogenic protein, can activate latent antiangiogenic and proangiogenic sites, respectively. In this study, we investigated the distribution of CgA-derived polypeptides in multiple myeloma patients and the subsequent implications for disease progression. We show that the ratio of pro/antiangiogenic forms of CgA is altered in multiple myeloma patients compared with healthy subjects and that this ratio is higher in BM plasma compared with peripheral plasma, suggesting enhanced local cleavage of the CgA C-terminal region. Enhanced cleavage correlated with increased VEGF and FGF2 BM plasma levels and BM microvascular density. Using the Vk*MYC mouse model of multiple myeloma, we further demonstrate that exogenously administered CgA was cleaved in favor of the proangiogenic form and was associated with increased microvessel density. Mechanistic studies revealed that multiple myeloma and proliferating endothelial cells can promote CgA C-terminal cleavage by activating the plasminogen activator/plasmin system. Moreover, cleaved and full-length forms could also counter balance the pro/antiangiogenic activity of each other in in vitro angiogenesis assays. These findings suggest that the CgA-angiogenic switch is activated in the BM of multiple myeloma patients and prompt further investigation of this CgA imbalance as a prognostic or therapeutic target. Cancer Res; 76(7); 1781-91. ©2016 AACR.


Subject(s)
Bone Marrow/pathology , Chromogranin A/genetics , Multiple Myeloma/genetics , Peptides/genetics , Animals , Female , Humans , Male , Mice , Middle Aged , Multiple Myeloma/pathology , Neovascularization, Pathologic
17.
Blood ; 127(16): 1987-97, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26825709

ABSTRACT

Hypoxia-inducible transcription factors (HIFs) regulate a wide array of adaptive responses to hypoxia and are often activated in solid tumors and hematologic malignancies due to intratumoral hypoxia and emerging new layers of regulation. We found that in chronic lymphocytic leukemia (CLL), HIF-1α is a novel regulator of the interaction of CLL cells with protective leukemia microenvironments and, in turn, is regulated by this interaction in a positive feedback loop that promotes leukemia survival and propagation. Through unbiased microarray analysis, we found that in CLL cells, HIF-1α regulates the expression of important chemokine receptors and cell adhesion molecules that control the interaction of leukemic cells with bone marrow and spleen microenvironments. Inactivation of HIF-1α impairs chemotaxis and cell adhesion to stroma, reduces bone marrow and spleen colonization in xenograft and allograft CLL mouse models, and prolongs survival in mice. Of interest, we found that in CLL cells, HIF-1α is transcriptionally regulated after coculture with stromal cells. Furthermore, HIF-1α messenger RNA levels vary significantly within CLL patients and correlate with the expression of HIF-1α target genes, including CXCR4, thus further emphasizing the relevance of HIF-1α expression to CLL pathogenesis.


Subject(s)
Cell Communication/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Tumor Microenvironment/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Adhesion/genetics , Chemotaxis, Leukocyte/genetics , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/metabolism , Spleen/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
18.
Exp Cell Res ; 330(1): 1-12, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25447443

ABSTRACT

Angiopoietin-2 (Ang-2) is involved in angiogenesis in both solid and hematological malignancies. In Multiple Myeloma (MM), serum Ang-2 correlates with disease progression and response to therapy. To address the patho-physiologic role of Ang-2 in MM associated angiogenesis, we used sera from patients with active MM, which contained significantly higher levels of the molecule, compared to those from patients with smoldering MM and Monoclonal Gammopathy of Undetermined Significance. MM Bone Marrow (BM) sera with high Ang-2 concentration specifically contributed to endothelial cell (EC) activation, while Ang-1 containing sera maintained EC stabilization. The functional dichotomy of Ang-1 and Ang-2 was confirmed by the triggering of distinctive signaling pathways down-stream the common Tie-2 receptor, i.e., the Akt or the ERK- phosphorylation pathway. Notably, Ang-2 but not VEGF serum levels correlated with BM micro-vessel density, further underscoring the key role of Ang-2 in angiogenesis. Western Blot, RT-PCR and immunocytochemistry identified MMEC as the major source of Ang-2, at variance with MM cells and CD14(+) BM monocytes. These data suggest that Ang-2 produced in the BM milieu may contribute to MM angiogenesis and suggest that the molecule can be further exploited both as angiogenesis biomarker and as a potential therapeutic target.


Subject(s)
Angiopoietin-2/metabolism , Bone Marrow/metabolism , Multiple Myeloma/metabolism , Neovascularization, Pathologic/metabolism , Adult , Aged , Aged, 80 and over , Angiopoietin-1/blood , Angiopoietin-1/metabolism , Angiopoietin-2/blood , Case-Control Studies , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MAP Kinase Signaling System , Male , Middle Aged , Multiple Myeloma/pathology , Receptor, TIE-2/metabolism , Vascular Endothelial Growth Factor A/blood
19.
Ann Rheum Dis ; 74(8): 1596-602, 2015 Aug.
Article in English | MEDLINE | ID: mdl-24671772

ABSTRACT

OBJECTIVES: Erdheim-Chester disease (ECD) is a rare form of histiocytosis characterised by uncontrolled chronic inflammation. The oncogenic BRAF(V600E) mutation has been reported in biopsies in 19 out of 37 patients with ECD from the largest published cohort, but never found in the patients' peripheral blood. Also, the role of the mutation in the pathogenesis of the disease has not been elucidated yet. BRAF(V600E) has been associated with oncogene-induced senescence (OIS), a protective mechanism against oncogenic events, characterised by the induction of proinflammatory pathways. METHODS: We verified the BRAF status in biopsies and peripheral blood from 18 patients with ECD from our cohort and matched controls by means of immunohistochemistry and of an ultrasensitive assay, based on the combination of a locked nucleic acid PCR and pyrosequencing. Droplet digital PCR was used to confirm the findings. We also evaluated the presence of senescence markers in ECD histiocytes. RESULTS: BRAF(V600E) mutation was present in all the biopsy and peripheral blood samples from patients with ECD and in none of the controls. ECD histiocytes and a fraction of circulating monocytes from patients with ECD showed signs of a constitutive activation of the MAPK pathway. Moreover, BRAF-mutated histiocytes expressed markers of OIS. CONCLUSIONS: The oncogenic BRAF(V600E) mutation is present in biopsies and in the peripheral blood from all patients with ECD who were evaluated and is associated with OIS. These findings have significant implications for the pathogenesis, diagnosis and treatment of ECD.


Subject(s)
Cellular Senescence/genetics , Erdheim-Chester Disease/genetics , Histiocytes/physiology , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Female , Histiocytes/metabolism , Humans , Immunohistochemistry , Inflammation/genetics , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Mutation , Oncogenes/physiology , Polymerase Chain Reaction/methods , Sequence Analysis
20.
Blood ; 124(4): 483-92, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24850756

ABSTRACT

Erdheim-Chester disease (ECD) is a rare, non-Langerhans histiocytosis. Recent findings suggest that ECD is a clonal disorder, marked by recurrent BRAFV600E mutations in >50% of patients, in which chronic uncontrolled inflammation is an important mediator of disease pathogenesis. Although ∼500 to 550 cases have been described in the literature to date, increased physician awareness has driven a dramatic increase in ECD diagnoses over the last decade. ECD frequently involves multiple organ systems and has historically lacked effective therapies. Given the protean clinical manifestations and the lack of a consensus-derived approach for the management of ECD, we provide here the first multidisciplinary consensus guidelines for the clinical management of ECD. These recommendations were outlined at the First International Medical Symposium for ECD, comprised of a comprehensive group of international academicians with expertise in the pathophysiology and therapy of ECD. Detailed recommendations on the initial clinical, laboratory, and radiographic assessment of ECD patients are presented in addition to treatment recommendations based on critical appraisal of the literature and clinical experience. These formalized consensus descriptions will hopefully facilitate ongoing and future research efforts in this disorder.


Subject(s)
Consensus , Erdheim-Chester Disease/diagnosis , Erdheim-Chester Disease/therapy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...