Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 257
Filter
1.
Clin Exp Immunol ; 198(2): 261-272, 2019 11.
Article in English | MEDLINE | ID: mdl-31165469

ABSTRACT

Primary Sjögren's syndrome (pSS) is a chronic inflammatory, autoimmune and systemic disorder commonly associated with dry eyes and a dry mouth. Recently, the hypothetical link between epithelial-mesenchymal transition (EMT)-dependent salivary gland (SG) fibrosis and chronic inflammatory conditions has been suggested. In this study, we present data demonstrating a negative correlation of the epithelial marker E-cadherin expression and a positive correlation of mesenchymal vimentin and collagen type I expression with increasing degrees of tissue inflammation in pSS SG specimens. In addition, as it is not clear whether dysregulated cytokines in pSS, interleukin (IL)-17 and IL-22 may also contribute to the EMT-dependent fibrosis process, the effect of IL-17 and IL-22 treatment on EMT-dependent SG fibrosis was evaluated in primary human salivary gland epithelial cells (SGEC) isolated from healthy subjects. Here we present data demonstrating that IL-17 and IL-22 can induce SGEC to undergo a morphological and phenotypical transition to a mesenchymal phenotype. In support of this, vimentin and collagen type I were up-regulated while a decreased expression of E-cadherin occurs after interleukin treatment, and co-operation between IL-17 and Il-22 was required to induce the EMT.


Subject(s)
Epithelial Cells/immunology , Epithelial-Mesenchymal Transition/immunology , Interleukin-17/immunology , Interleukins/immunology , Salivary Glands/immunology , Sjogren's Syndrome/immunology , Aged , Antigens, CD/immunology , Cadherins/immunology , Collagen Type I/immunology , Epithelial Cells/pathology , Female , Humans , Inflammation/immunology , Inflammation/pathology , Male , Middle Aged , Salivary Glands/pathology , Sjogren's Syndrome/pathology , Up-Regulation/immunology , Vimentin/immunology , Interleukin-22
2.
J Plast Reconstr Aesthet Surg ; 72(9): 1509-1517, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31202697

ABSTRACT

INTRODUCTION: Finger degloving injuries (FDIs) represent a challenge in hand surgery. When replantation is not possible, several techniques including loco-regional flaps, pocket abdominal flaps and free flaps have been described as methods to provide skin cover and avoid finger shortening. The aim of this study is to present our experience with acellular dermal matrices (ADMs) in the treatment of FDI. MATERIALS AND METHODS: We retrospectively reviewed the charts of 18 patients who presented with FDI and were treated with ADM between December 2015 and July 2017. Surgical outcomes including complications were analysed, and patient-centred assessments were performed at 12 months of follow-up. RESULTS: The follow-up period ranged from 10 to 20 months. All patients showed good integration and vascularisation of the ADM. All the fingers covered with ADM were firm and soft, with a slim and satisfactory appearance at a mean follow-up of 12 months. No limitations in tendon sliding were observed at dynamic sonography one year after surgery. CONCLUSION: ADMs could be regarded as a viable option when dealing with FDIs, if replantation is not possible and finger length is to be preserved. On the basis of these results, the surgical treatment of FDI with ADM is a viable option that produces good functional outcomes and cosmetic appearance.


Subject(s)
Acellular Dermis , Degloving Injuries/surgery , Finger Injuries/surgery , Free Tissue Flaps , Plastic Surgery Procedures/methods , Range of Motion, Articular/physiology , Skin Transplantation/methods , Adult , Aged , Biopsy , Degloving Injuries/diagnosis , Degloving Injuries/physiopathology , Female , Finger Injuries/diagnosis , Finger Injuries/physiopathology , Follow-Up Studies , Humans , Male , Middle Aged , Retrospective Studies , Time Factors , Treatment Outcome , Ultrasonography , Young Adult
3.
Carbohydr Polym ; 198: 124-130, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30092982

ABSTRACT

We aimed at producing a hydrogel from a chitosan (CS) derivative soluble in physiological conditions to avoid any purification step thus allowing to use the materials also as an in-situ forming material. So, we crosslinked glycol chitosan (GCS) with poly(ethylene glycol) diglycidyl ether (PEGDE) in water at 37 °C. The scaffolds, referred as GCS-PEG, were specifically designed to be used as wound dressing materials as such (after crosslinking) or as in-situ forming materials. Different amounts of PEGDE were tested. The obtained scaffolds showed macroscopic pores and a tailorable swelling in water by controlling the crosslinking degree. Moreover, GCS-PEG scaffolds displayed a significant antimicrobial activity against Staphylococcus aureus. In-vivo study using the chick embryo choriallantoic membrane resulted in a highly pronounced pro-angiogenic activity suggesting important tissue regeneration properties. Moreover, the employed materials are commercially available, no organic solvents are required and the scaling up is quite predictable.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chitosan/pharmacology , Epoxy Resins/pharmacology , Hydrogels/pharmacology , Neovascularization, Physiologic/drug effects , Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/chemistry , Chick Embryo , Chitosan/chemistry , Epoxy Resins/chemistry , Hydrogels/chemistry , Staphylococcus aureus/growth & development
4.
Thromb Res ; 140 Suppl 1: S182, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27161705

ABSTRACT

INTRODUCTION: Platelet thrombospondin-1 (TSP-1) is a major endogenous regulator of growth factor activity in physiological and pathological processes, including tumor onset, progression and angiogenesis. We previously demonstrated that TSP-1 binds to FGF-2, sequestering the growth factor and inhibiting its angiogenic activity. We also identified a non-peptidic antiangiogenic compound (SM27) that retains the structural and functional properties of the FGF2-binding sequence of TSP-1. AIM: To identify new small molecule inhibitors of FGF2 that recapitulate the structure and functional properties of the FGF-2-binding site of TSP-1, by investigating the chemical space around SM27. MATERIALS AND METHODS: A similarity-based screening of small molecule libraries has been used to identify candidates, followed by docking calculations, and evaluation of the activity of the resulting compounds in biochemical and biophysical assays, to assess interaction with FGF2, and in experimental models of angiogenesis, to assess biological activity. RESULTS: The used integrated approach allowed selecting 7 bi-naphthalenic compounds that bound FGF2 inhibiting FGF2 binding to both heparan sulfate proteoglycans and FGFR1. The compounds inhibited FGF2-induced endothelial cell proliferation, vessel sprouting from aortic rings and angiogenesis in the chorioallantoic membrane assay, with improved potency over SM27. CONCLUSIONS: We have identified new compounds that are valuable as FGF inhibitors for potential therapeutic purposes. Moreover, these compounds are useful chemical tools to identify the minimal stereochemical requirements for FGF2 binding and activity to improve the design of new agents for antineoplastic therapy. ACKNOWLEDGEMENT: Supported by AIRC (Associazione Italiana per la Ricerca sul Cancro).

6.
Leukemia ; 30(3): 640-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26487273

ABSTRACT

Bortezomib (bort) has improved overall survival in patients with multiple myeloma (MM), but the majority of them develop drug resistance. In this study, we demonstrate that bone marrow (BM) fibroblasts (cancer-associated fibroblasts; CAFs) from bort-resistant patients are insensitive to bort and protect the RPMI8226 and patients' plasma cells against bort-induced apoptosis. Bort triggers CAFs to produce high levels of interleukin (IL)-6, IL-8, insulin-like growth factor (IGF)-1 and transforming growth factor (TGF) ß. Proteomic studies on CAFs demonstrate that bort resistance parallels activation of oxidative stress and pro-survival autophagy. Indeed, bort induces reactive oxygen species in bort-resistant CAFs and activates autophagy by increasing light chain 3 protein (LC3)-II and inhibiting p62 and phospho-mammalian target of rapamycin. The small-interfering RNA knockdown of Atg7, and treatment with 3-methyladenine, restores bort sensitivity in bort-resistant CAFs and produces cytotoxicity in plasma cells co-cultured with CAFs. In the syngeneic 5T33 MM model, bort-treatment induces the expansion of LC3-II(+) CAFs. TGFß mediates bort-induced autophagy, and its blockade by LY2109761, a selective TßRI/II inhibitor, reduces the expression of p-Smad2/3 and LC3-II and induces apoptosis in bort-resistant CAFs. A combination of bort and LY2109761 synergistically induces apoptosis of RPMI8226 co-cultured with bort-resistant CAFs. These data define a key role for CAFs in bort resistance of plasma cells and provide the basis for a novel targeted therapeutic approach.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Multiple Myeloma/drug therapy , Pyrazoles/pharmacology , Pyrroles/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Aged , Aged, 80 and over , Animals , Autophagy/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Drug Combinations , Drug Resistance, Neoplasm/genetics , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Plasma Cells/drug effects , Plasma Cells/metabolism , Plasma Cells/pathology , Primary Cell Culture , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Survival Analysis , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
7.
Leukemia ; 29(4): 958-67, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25283844

ABSTRACT

Interleukin (IL)-31A binds to an heterodimer composed of IL-31 receptor A (IL-31RA) and Oncostatin M Receptor (OSMR). The IL-31/IL-31R complex is involved in the pathogenesis of various skin diseases, including cutaneous T-cell lymphoma. No information is available on the relations between the IL-31/IL-31R complex and B-cell lymphoma. Here we have addressed this issue in follicular lymphoma (FL), a prototypic germinal center(GC)-derived B-cell malignancy. IL-31 enhanced primary FL cell proliferation through IL-31R-driven signal transducer and activator of transcription factor 1/3 (STAT1/3), extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt phosphorylation. In contrast, GC B cells did not signal to IL-31 in spite of IL-31R expression. GC B cells expressed predominantly the inhibitory short IL-31RA isoform, whereas FL cells expressed predominantly the long signaling isoform. Moreover, GC B cells lacked expression of other IL-31RA isoforms potentially involved in the signaling pathway. IL-31 protein expression was significantly higher in surface membrane than in cytosol of both FL and GC B cells. IL-31 was detected in plasma membrane microvesicles from both cell types but not released in soluble form in culture supernatants. IL-31 and IL-31RA expression was higher in lymph nodes from FL patients with grade IIIa compared with grade I/II, suggesting a paracrine and/or autocrine role of IL-31/IL-31RA complex in tumor progression through microvesicle shedding.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Leukemic , Germinal Center/metabolism , Interleukins/genetics , Lymphoma, Follicular/genetics , Receptors, Interleukin/genetics , B-Lymphocytes/pathology , Cell Membrane/metabolism , Cell Proliferation , Cell-Derived Microparticles/metabolism , Cytosol/metabolism , Female , Germinal Center/pathology , Humans , Interleukins/metabolism , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Grading , Phosphorylation , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
8.
Leukemia ; 28(4): 904-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23995611

ABSTRACT

The role of cancer-associated fibroblasts (CAFs) has not been previously studied in multiple myeloma (MM). Here, cytofluorimetric analysis revealed higher proportions of bone marrow (BM) CAFs in patients with active MM (both at diagnosis and relapse) compared with patients in remission or those with monoclonal gammopathy of undetermined significance or deficiency anemia (controls). CAFs from MM patients produced increased levels of transforming growth factor-ß, interleukin-6, stromal cell-derived factor-1α, insulin-like growth factor-1, vascular endothelial growth factor and fibroblast growth factor-2 and displayed an activated and heterogeneous phenotype, which supported their origin from resident fibroblasts, endothelial cells and hematopoietic stem and progenitor cells via the endothelial-mesenchymal transition as well as mesenchymal stem cells via the mesenchymal transition, as both of these processes are induced by MM cells and CAFs. Active MM CAFs fostered chemotaxis, adhesion, proliferation and apoptosis resistance in MM cells through cytokine signals and cell-to-cell contact, which were inhibited by blocking CXCR4, several integrins and fibronectin. MM cells also induced the CAFs proliferation. In syngeneic 5T33MM and xenograft mouse models, MM cells induced the expansion of CAFs, which, in turn, promoted MM initiation and progression as well as angiogenesis. In BM biopsies from patients and mice, nests of CAFs were found in close contact with MM cells, suggesting a supportive niche. Therefore, the targeting of CAFs in MM patients may be envisaged as a novel therapeutic strategy.


Subject(s)
Bone Marrow Cells/physiology , Fibroblasts/physiology , Multiple Myeloma/pathology , Aged , Aged, 80 and over , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Chemokine CXCL12/physiology , Disease Progression , Epithelial-Mesenchymal Transition , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype
9.
Crit Rev Oncol Hematol ; 88(1): 187-97, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23561333

ABSTRACT

Murine cancer models have been extremely useful for analyzing the biology of pathways involved in cancer initiation, promotion, and progression. Interestingly, several murine cancer models also exhibit heterogeneity, genomic instability and an intact immune system. However, they do not adequately represent several features that define cancer in humans, including long periods of latency, the complex biology of cancer recurrence and metastasis and outcomes to novel therapies. Therefore, additional models that better investigate the human disease are needed. In the pet population, with special references to the dog, cancer is a spontaneous disease and dogs naturally develop cancers that share many characteristics with human malignancies. More than 40 years ago, optimization of bone marrow transplantation protocols was undertaken in dogs and recently novel targeted therapies such as liposomal muramyl tripeptide phosphatidylethanolamine and several tyrosine kinase inhibitors, namely masitinib (AB1010) and toceranib phosphate (SU11654), have been developed to treat dog tumors which have then been translated to human clinical trials. In this review article, we will analyze biological data from dog tumors and comparative features with human tumors, and new therapeutic approaches translated from dog to human cancer.


Subject(s)
Neoplasms/etiology , Animals , Disease Models, Animal , Dogs , Humans , Neoplasms/diagnosis , Neoplasms/therapy , Translational Research, Biomedical
10.
Genes Immun ; 13(5): 411-20, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22513453

ABSTRACT

We explore the involvement of tumor necrosis factor α (TNF-α)-converting enzyme (TACE) in vascular endothelial growth factor (VEGF) and its receptor 2 (VEGFR2) (VEGF-A/VEGFR2)-mediated angiogenesis in Sjögren's syndrome (SS), one of the most common autoimmune rheumatic diseases. To test the hypothesis that SS autoantibodies (Abs) regulate VEGF-A/VEGFR2 expression by a TACE-dependent nuclear factor-κB (NF-κB) activation pathway, their effects on the expression and activation of the VEGF-A/TACE/VEGFR2/NF-κB pathway were determined in human salivary gland epithelial cells (SGEC). An enhanced angiogenesis in SS salivary gland biopsies was observed, associated with an increased VEGF-A expression and activation of VEGF-A/VEGFR2 signaling. Human cytokine array analysis of the pro-inflammatory and pro-angiogenic protein response in SGEC treated with SS Abs revealed an overexpression of multiple pro-angiogenic factors. TACE RNA knockdown, the use of anti-VEGF-A monoclonal antibody and the inhibition of NF-κB activity significantly abrogated the release of pro-angiogenic factors, demonstrating that VEGF-A/TACE/VEGFR2/NF-κB axis dysfunction may be contributory to pathogenesis and exacerbation of this autoimmune condition.


Subject(s)
ADAM Proteins/metabolism , NF-kappa B/metabolism , Neovascularization, Pathologic/metabolism , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Adult , Aged , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , Autoantibodies/pharmacology , Case-Control Studies , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/pathology , Cytokines/metabolism , Enzyme Activation/drug effects , Female , Humans , Male , Middle Aged , Signal Transduction/drug effects , Sjogren's Syndrome/genetics
11.
Curr Med Chem ; 19(7): 955-60, 2012.
Article in English | MEDLINE | ID: mdl-22214461

ABSTRACT

The immune system regulates angiogenesis in cancer by means of pro- and anti-angiogenesis activities. In fact, both innate (macrophages, granulocytes, mast cells, dendritic cells, natural killer cells, and platelets) and adaptive (T and B lymphocytes) immune cells synthesize several pro- and anti-angiogenic mediators. Moreover, in pre-clinical models, a synergy has been observed between antiangiogenic molecules and immunotherapy. In this review article, we will focus on some angiogenenic and anti-angiogenic molecules properties of immune cells that may be utilized for a potential parmacological use as anti-angiogenic agents in cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/drug therapy , Adaptive Immunity , Humans , Immunity, Innate , Inflammation , Neoplasms/immunology
12.
Oncogene ; 31(18): 2258-69, 2012 May 03.
Article in English | MEDLINE | ID: mdl-21963844

ABSTRACT

Bone marrow (BM) angiogenesis has an important role in the initiation and progression of multiple myeloma (MM). We looked at novel mechanisms of vessel formation in patients with MM through a comparative proteomic analysis between BM endothelial cells (ECs) of patients with active MM (MMECs) and ECs of patients with monoclonal gammopathy of undetermined significance (MGECs) and of subjects with benign anemia (normal ECs). Four proteins were found overexpressed in MMECs: filamin A, vimentin, α-crystallin B and 14-3-3ζ/δ protein, not yet linked to overangiogenic phenotype. These proteins gave a typical distribution in the BM of MM patients and in MMECs versus MGECs, plausibly according to a different functional state. Their expression was enhanced by vascular endothelial growth factor, fibroblast growth factor 2, hepatocyte growth factor and MM plasma cell conditioned medium in step with enhancement of MMEC angiogenesis. Their silencing RNA knockdown affected critical MMEC angiogenesis-related functions, such as spreading, migration and tubular morphogenesis. A gradual stabilization of 14-3-3ζ/δ protein was observed, with transition from normal ECs to MGECs and MMECs that may be a critical step for the angiogenic switch in MMECs and maintenance of the cell overangiogenic phenotype. These proteins were substantially impacted by anti-MM drugs, such as bortezomib, lenalidomide and panobinostat. Results suggest that these four proteins could be new targets for the antiangiogenic management of MM patients.


Subject(s)
Multiple Myeloma/blood supply , Multiple Myeloma/pathology , Neovascularization, Pathologic/genetics , 14-3-3 Proteins/genetics , Aged , Aged, 80 and over , Anemia/genetics , Anemia/pathology , Bone Marrow Cells/pathology , Cell Movement , Contractile Proteins/genetics , Endothelial Cells/pathology , Female , Filamins , Humans , Male , Microfilament Proteins/genetics , Middle Aged , Molecular Targeted Therapy , Multiple Myeloma/genetics , Paraproteinemias/genetics , Paraproteinemias/pathology , Proteomics , Vimentin/genetics , alpha-Crystallins/genetics
13.
Leukemia ; 26(2): 225-35, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21844875

ABSTRACT

Childhood acute myeloid leukemia (AML) is a hematological malignancy in which tumor burden is continuously replenished by leukemic-initiating cells (ICs), which proliferate slowly and are refractory to chemotherapeutic agents. We investigated whether interleukin (IL)-12, an immuno-modulatory cytokine with anti-tumor activity, may target AML blasts (CD45(+)CD33(+)) and populations known to contain leukemia ICs (that is, CD34(+)CD38(-), CD33(+)CD38(+) and CD44(+)CD38(-) cells). We demonstrate for the first time that: i) AML blasts and their CD34(+)CD38(-), CD33(+)CD38(+), CD44(+)CD38(-) subsets express the heterodimeric IL-12 receptor (IL-12R), ii) AML cells injected subcutaneously into NOD/SCID/Il2rg(-/-) (NSG) mice developed a localized tumor mass containing leukemic ICs and blasts that were virtually eliminated by IL-12 treatment, iii) AML cells injected intravenously into NSG mice engrafted within the first month in the spleen, but not in bone marrow or peripheral blood. At this time, IL-12 dramatically dampened AML CD45(+)CD33(+), CD34(+)CD38(-), CD33(+)CD38(+) and CD44(+)CD38(-) populations, only sparing residual CD33(+)CD38(+) cells that did not express IL-12Rß2. From 30 to 60 days after the initial inoculum, these IL-12-unresponsive cells expanded and metastasized in both control and IL-12-treated NSG mice. Our data indicate that the absence of IL-12Rß2 in pediatric AML cells favours leukemia progression in NOD/SCID/IL2Rγc-deficient mice.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Interleukin-12 Receptor beta 2 Subunit/metabolism , Leukemia, Myeloid, Acute/pathology , Adolescent , Adult , Animals , Cell Division , Child , Child, Preschool , Disease Progression , Female , Flow Cytometry , Humans , Infant , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Real-Time Polymerase Chain Reaction , Sialic Acid Binding Ig-like Lectin 3
14.
Leukemia ; 26(6): 1365-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22193967

ABSTRACT

Interleukin (IL)-23 and IL-27 are pro-inflammatory cytokines that share functional and structural similarities and may exert anti-tumor activities against solid and hematological malignancies. Here, we asked whether IL-23 and IL-27, alone or in combination, may act directly against human follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL) cells. In this study, we demonstrated for the first time that human primary FL and DLBCL cells expressed complete and functional IL-23 and IL-27 receptors (R) and that IL-23 and IL-27 exerted anti-tumor activities in vitro and in vivo through different and complementary mechanisms. In vivo studies using severe combined immunodeficiency /non-obese diabetic mice-injected subcutaneously with human SU-DHL-4 cell line revealed that IL-23 inhibited directly tumor-cell proliferation, whereas IL-27 impaired the angiogenic program of lymphoma cells resulting in strong reduction of cell growth. In addition, combined treatment of IL-23 and IL-27 amplified the anti-tumor effects in vivo as compared with administration of each cytokine alone. These anti-tumor mechanisms were confirmed by in vitro experiments performed with primary lymphoma cells and cell lines. Our results strongly encourage the development of future clinical trials to evaluate the toxicity and efficacy of the IL-23 and IL-27 in lymphoma patients.


Subject(s)
Interleukin-17/therapeutic use , Interleukin-23/therapeutic use , Lymphoma, Follicular/prevention & control , Lymphoma, Large B-Cell, Diffuse/prevention & control , Aged , Aged, 80 and over , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Chickens , Chorioallantoic Membrane , DNA, Neoplasm/genetics , Drug Synergism , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Rom J Morphol Embryol ; 52(3 Suppl): 981-4, 2011.
Article in English | MEDLINE | ID: mdl-22119813

ABSTRACT

The aim of this study was to evaluate the role of macrophages and mast cells and of microvascular density in atherosclerotic plaques collected from 63 consecutive symptomatic and asymptomatic patients undergoing carotid endarterectomy for carotid disease. Results have shown no statistically significant differences between the two groups as concerns: (i) the degree of stenosis; (ii) the extention of the lipidic core; (iii) the thickness of the fibrous cup; (iv) the inflammatory infiltrate; (v) the degree of calcification; (vi) the intraplaque hemorrhage. Otherwise, statistically significant difference was found in microvascular density, in the number of CD68-positive macrophages and tryptase-positive mast cells in plaques from symptomatic patients, as compared to asymptomatic patients. Overall, this study indicate that although advanced symptomatic and asymptomatic carotid plaques present similar histomorphological characteristics, the degree of macrophage and mast cell infiltration and differences in microvascular density could help to discriminate between symptomatic and asymptomatic patients.


Subject(s)
Carotid Arteries/pathology , Macrophages/pathology , Mast Cells/pathology , Plaque, Atherosclerotic/pathology , Biopsy , Female , Humans , Male , Microvessels/pathology
16.
Leukemia ; 25(12): 1815-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21701492

ABSTRACT

B-acute lymphoblastic leukemia (B-ALL) represents the most common pediatric hematological tumor that derives from the aberrant proliferation of early B lymphocytes in the bone marrow. Although most of the B-ALL children take advantage from current therapeutic protocols, some patients relapse and need alternative therapies. With this background, we investigated whether interleukin (IL)-27, an immunomodulatory cytokine with antitumor properties, may function as an antitumor agent against pediatric B-ALL cells. Here we show for the first time that pediatric B-ALL cells functional IL-27R and that IL-27 dampens directly tumor growth in vivo and in vitro through mechanisms elucidated in this study. The novelty of these results deals with the first demonstration that (1) B-ALL cells from pediatric patients injected intravenously (i.v.) into NOD/SCID/Il2rg(-/-) (NSG) mice gave rise to leukemic spreading that was severely hampered by IL-27; (2) IL-27-treated mice, compared with controls, showed significant reduction of putative B-ALL-initiating cells and blasts in the peripheral blood (PB), bone marrow (BM) and spleen; and that (3) IL-27 reduced in vitro B-ALL cell proliferation and angiogenesis, induced apoptosis and downregulated miR-155. Our results strongly encourage the development of future clinical trials to evaluate the toxicity and efficacy of IL-27 in childhood B-ALL patients.


Subject(s)
Apoptosis , Interleukin-17/therapeutic use , Leukemia, B-Cell/pathology , Leukemia, B-Cell/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Adolescent , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Chickens , Child , Child, Preschool , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Female , Flow Cytometry , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Infant , Leukemia, B-Cell/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
17.
Leukemia ; 25(8): 1268-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21546901

ABSTRACT

Several chemokines/chemokine receptors such as CCR7, CXCR4 and CXCR5 attract chronic lymphocytic leukemia (CLL) cells to specific microenvironments. Here we have investigated whether the CX(3)CR1/CX(3)CL1 axis is involved in the interaction of CLL with their microenvironment. CLL cells from 52 patients expressed surface CX(3)CR1 and CX(3)CL1 and released constitutively soluble CX(3)CL1. One third of these were attracted in vitro by soluble CX(3)CL1. CX(3)CL1-induced phosphorylation of PI3K, Erk1/2, p38, Akt and Src was involved in induction of CLL chemotaxis. Leukemic B cells upregulated CXCR4 upon incubation with CX(3)CL1 and this was paralleled by increased chemotaxis to CXCL12. Akt phosphorylation was involved in CX(3)CL1-induced upregulation of CXCR4 on CLL. In proliferation centers from CLL lymph node and bone marrow, CX(3)CL1 was expressed by CLL cells whereas CX(3)CR1 was detected in CLL and stromal cells. Nurselike cells (NLCs) generated from CLL patient blood co-expressed surface CX(3)CR1 and CX(3)CL1, but did not secrete soluble CX(3)CL1. Only half of NLC cell fractions were attracted in vitro by CX(3)CL1. In conclusion, the CX(3)CR1/CX(3)CL1 system may contribute to interactions between CLL cells and tumor microenvironment by increasing CXCL12-mediated attraction of leukemic cells to NLC and promoting directly adhesion of CLL cells to NLC.


Subject(s)
Cell Communication , Chemokine CX3CL1/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Receptors, Chemokine/physiology , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , CX3C Chemokine Receptor 1 , Chemotaxis , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymph Nodes/metabolism , Male , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
19.
Leukemia ; 25(3): 527-37, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21183939

ABSTRACT

The deregulation of the homeobox genes as homeoboxB (HOXB)-7 has been previously associated to tumor progression and angiogenesis; here we investigated the potential role of HOXB7 in the pro-angiogenic properties of multiple myeloma (MM) cells. We found that HOXB7 was expressed in 10 out of 22 MM patients analyzed at the diagnosis related to high bone marrow angiogenesis and overexpressed in about 40% of myeloma cell lines compared with normal plasma cells. Enforced HOXB7 expression in MM cells by a lentiviral vector significantly modified their transcriptional and angiogenic profile, checked by combined microarray and angiogenesis PCR analyses, upregulating VEGFA, FGF2, MMP2, WNT5a and PDGFA and downregulating thrombospoindin-2. The pro- and anti-angiogenic HOXB7-related gene signature was also validated in a large independent dataset of MM patients. Accordingly, MM-induced vessel formation was significantly increased by HOXB7 overexpression both in vitro angiogenic and chorioallantoic membrane assays, as well as the HOXB7 silencing by small interfering RNA inhibited the production of angiogenic factors, and the pro-angiogenic properties of MM cells. Finally, in SCID-NOD mice we confirmed that HOXB7 overexpression by MM cells stimulated tumor growth, increased MM-associated angiogenesis and the expression of pro-angiogenic genes by microarray analysis supporting the critical role of HOXB7 in the angiogenic switch in MM.


Subject(s)
Homeodomain Proteins/physiology , Multiple Myeloma/blood supply , Neovascularization, Pathologic/etiology , Aged , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...