Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Adv Exp Med Biol ; 1408: 273-290, 2023.
Article in English | MEDLINE | ID: mdl-37093433

ABSTRACT

The Philadelphia-negative myeloproliferative neoplasms (MPNs), defined as clonal disorders of the hematopoietic stem cells, are characterized by the proliferation of mature myeloid cells in the bone marrow and a chronic inflammatory status impacting the initiation, progression, and symptomatology of the malignancies. There are three main entities defined as essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF), and genetically classified by JAK2V617F, CALR, or MPL mutations. In MPNs, due to the overproduction of inflammatory cytokines by the neoplastic cells and non-transformed immune cells, chronic inflammation may provoke the generation and expansion of myeloid-derived suppressors cells (MDSCs) that highly influence the adaptive immune response. Although peripheral blood MDSC levels are elevated, their frequency in the bone marrow of MPNs patients is not well elucidated yet. Our results indicated increased levels of total (T)-MDSCs (CD33+HLA-DR-/low) and polymorphonuclear (PMN)-MDSCs (CD33+/HLA-DRlow/CD15+/CD14-) in the bone marrow and peripheral blood of all three types of MPNs malignancies. However, these bone marrow MDSCs-increased frequencies did not correlate with the clinical parameters, such as hepatomegaly, leukocytes, hemoglobin, or platelet levels, or with JAK2 and CALR mutations. Besides, bone marrow MDSCs, from ET, PV, and PMF patients, exhibited immunosuppressive function, determined as T-cell proliferation inhibition. Notably, the highest T-MDSCs and PMN-MDSC levels were found in PMF samples, and the increased MDSCs frequency strongly correlated with the degree of myelofibrosis. Thus, these data together indicate that the immunosuppressive MDSCs population is increased in the bone marrow of MPNs patients and may be implicated in generating a fibrotic microenvironment.


Subject(s)
Myeloid-Derived Suppressor Cells , Myeloproliferative Disorders , Polycythemia Vera , Primary Myelofibrosis , Thrombocythemia, Essential , Humans , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Bone Marrow/pathology , Myeloid-Derived Suppressor Cells/pathology , Myeloproliferative Disorders/genetics , Polycythemia Vera/genetics , Polycythemia Vera/pathology , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/pathology , Janus Kinase 2/genetics , Mutation , Tumor Microenvironment
2.
Int J Mol Sci ; 24(14)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37511129

ABSTRACT

Psychological stress is a significant contributor to various chronic diseases and affects multiple physiological processes including erythropoiesis. This study aimed to examine the tissue-specific contributions of macrophages and extracellular ATP, as a signal of disturbed tissue homeostasis, to erythropoiesis under conditions of repeated psychological stress. Adult male BALB/c mice were subjected to 2 h daily restraint stress for seven consecutive days. Clodronate-liposomes were used to deplete resident macrophages from the bone marrow and spleen two days prior to the first restraint procedure, as well as newly recruited macrophages, every third day for the duration of the experiment. Repeated stress induced a considerable increase in the number of erythroid progenitor cells as well as in the percentage of CD71+/Ter119+ and CD71-/Ter119+ cells in the bone marrow and spleen. Macrophage depletion completely abolished the stimulative effect of repeated stress on immature erythroid cells, and prevented stress-induced increases in ATP levels, P2X7 receptor (P2X7R) expression, and ectonucleotidase CD39 activity and expression in the bone marrow and spleen. The obtained results demonstrate the stimulative effects of repeated stress on erythroid cells, extracellular ATP levels, P2X7R expression, CD39 activity and expression within the bone marrow and spleen, as well as the essential role of macrophages in stress-induced changes.


Subject(s)
Erythropoiesis , Macrophages , Mice , Animals , Male , Macrophages/metabolism , Spleen/metabolism , Mice, Inbred BALB C , Stress, Psychological , Adenosine Triphosphate/metabolism
3.
Dev Dyn ; 251(1): 105-124, 2022 01.
Article in English | MEDLINE | ID: mdl-33797140

ABSTRACT

Transforming growth factor-beta1 (TGF-ß1) plays a crucial role in tumor progression. It can inhibit early cancer stages but promotes tumor growth and development at the late stages of tumorigenesis. TGF-ß1 has a potent immunosuppressive function within the tumor microenvironment that largely contributes to tumor cells' immune escape and reduction in cancer immunotherapy responses. Likewise, myeloid-derived suppressor cells (MDSCs) have been postulated as leading tumor promoters and a hallmark of cancer immune evasion mechanisms. This review attempts to analyze the prominent roles of both TGF-ß1 and MDSCs and their interplay in cancer immunity. Furthermore, therapies against either TGF-ß1 or MDSCs, and their potential synergistic combination with immunotherapies are discussed. Simultaneous TGF-ß1 and MDSCs inhibition suggest a potential improvement in immunotherapy or subverted tumor immune resistance.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Humans , Myeloid-Derived Suppressor Cells/pathology , Neoplasms/pathology , Neoplasms/therapy , Transforming Growth Factor beta1 , Tumor Escape , Tumor Microenvironment
4.
Medicina (Kaunas) ; 59(10)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37893561

ABSTRACT

Background and Objectives: Oral squamous cell carcinoma (OSCC) accounts for about 95% of oral cancers. It represents a serious public health problem due to the high degree of morbidity and mortality, as well as multifactorial etiology. Human papillomavirus (HPV) infection is a well-documented risk factor for oropharyngeal carcinoma, but its role in oral carcinogenesis is still debatable. Our aim was to investigate the differences in the prevalence of high-risk HPV genotypes (HR-HPV) in patients with OSCC and oral potentially malignant disorders (OPMD) from that of healthy subjects. Materials and Methods: A total of 90 subjects were included in the cross-sectional study and divided into three groups of 30 patients each: (1) patients with OSCC, (2) patients with OPMD, and (3) healthy subjects. We examined the presence of 12 HR-HPV genotypes in the obtained biological material (oral swabs) using real-time PCR. Results: One or more of the 12 tested HR-HPV genotypes were detected in 5/30 patients with OSCC and 2/30 with OPMD, whereas no healthy subjects were positive for any of the tested genotypes. There was a statistically significant difference in nodal involvement between HPV-positive and HPV-negative patients with OSCC. Conclusions: Oral HR-HPV was detected in patients with oral premalignant and malignant lesions but not in healthy individuals, suggesting a possible role in oral carcinogenesis. Broad HR-HPV panel testing could increase the sensitivity of risk assessment and screening for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Diseases , Mouth Neoplasms , Papillomavirus Infections , Humans , Mouth Neoplasms/pathology , Pilot Projects , Human Papillomavirus Viruses , Carcinoma, Squamous Cell/pathology , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Serbia/epidemiology , Cross-Sectional Studies , Squamous Cell Carcinoma of Head and Neck , Papillomaviridae/genetics , Carcinogenesis
5.
Int J Mol Sci ; 23(15)2022 Aug 02.
Article in English | MEDLINE | ID: mdl-35955703

ABSTRACT

The increasing cancer incidence has certified oncological management as one of the most critical challenges for the coming decades. New anticancer strategies are still needed, despite the significant advances brought to the forefront in the last decades. The most recent, promising therapeutic approaches have benefitted from the application of human perinatal derivatives (PnD), biological mediators with proven benefits in several fields beyond oncology. To elucidate preclinical results and clinic outcomes achieved in the oncological field, we present a narrative review of the studies resorting to animal models to assess specific outcomes of PnD products. Recent preclinical evidence points to promising anticancer effects offered by PnD mediators isolated from the placenta, amniotic membrane, amniotic fluid, and umbilical cord. Described effects include tumorigenesis prevention, uncontrolled growth or regrowth inhibition, tumor homing ability, and adequate cell-based delivery capacity. Furthermore, PnD treatments have been described as supportive of chemotherapy and radiological therapies, particularly when resistance has been reported. However, opposite effects of PnD products have also been observed, offering support and trophic effect to malignant cells. Such paradoxical and dichotomous roles need to be intensively investigated. Current hypotheses identify as explanatory some critical factors, such as the type of the PnD biological products used or the manufacturing procedure to prepare the tissue/cellular treatment, the experimental design (including human-relevant animal models), and intrinsic pathophysiological characteristics. The effective and safe translation of PnD treatments to clinical practice relies on the collaborative efforts of all researchers working with human-relevant oncological preclinical models. However, it requires proper guidelines and consensus compiled by experts and health workers who accurately describe the methodology of tissue collection, PnD isolation, manufacturing, preservation, and delivery to the final user.


Subject(s)
Neoplasms , Animals , Female , Humans , Neoplasms/drug therapy , Pregnancy
6.
J Cell Physiol ; 236(11): 7322-7341, 2021 11.
Article in English | MEDLINE | ID: mdl-33934350

ABSTRACT

Mesenchymal stem cells (MSCs) have been identified within dental pulp tissues of exfoliated deciduous (SHEDs) and permanent (DPSCs) teeth. Although differences in their proliferative and differentiation properties were revealed, variability in SHEDs and DPSCs responsiveness to growth factors and cytokines have not been studied before. Here, we investigated the influence of interleukin-17 (IL-17) and basic fibroblast growth factor (bFGF) on stemness features of SHEDs and DPSCs by analyzing their proliferation, clonogenicity, cell cycle progression, pluripotency markers expression and differentiation after 7-day treatment. Results indicated that IL-17 and bFGF differently affected SHEDs and DPSCs proliferation and clonogenicity, since bFGF increased proliferative and clonogenic potential of both cell types, while IL-17 similarly affected SHEDs, exerting no effects on adult counterparts DPSCs. In addition, both factors stimulated NANOG, OCT4, and SOX2 pluripotency markers expression in SHEDs and DPSCs showing diverse intracellular expression patterns dependent on MSCs type. As for the differentiation capacity, both factors displayed comparable effects on SHEDs and DPSCs, including stimulatory effect of IL-17 on early osteogenesis in contrast to the strong inhibitory effect showed for bFGF, while having no impact on SHEDs and DPSCs chondrogenesis. Moreover, bFGF combined with IL-17 reduced CD90 and stimulated CD73 expression on both types of MSCs, whereas each factor induced IL-6 expression indicating its' role in IL-17/bFGF-modulated properties of SHEDs and DPSCs. All these data demonstrated that dental pulp MSCs from primary and permanent teeth exert intrinsic features, providing novel evidence on how IL-17 and bFGF affect stem cell properties important for regeneration of dental pulp at different ages.


Subject(s)
Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dental Pulp/drug effects , Fibroblast Growth Factor 2/pharmacology , Interleukin-17/pharmacology , Mesenchymal Stem Cells/drug effects , Tooth Exfoliation , Tooth, Deciduous/drug effects , Adult , Cells, Cultured , Child , Chondrogenesis/drug effects , Dental Pulp/cytology , Dental Pulp/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Phenotype , Tooth, Deciduous/cytology , Tooth, Deciduous/metabolism , Young Adult
7.
Eur Biophys J ; 50(6): 829-846, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33813598

ABSTRACT

Flow cytometry (FC) analysis of erythrocyte shape and related biomechanical properties, such as osmotic fragility, have not moved from a research tool to regular clinical testing. The main reason is existing evidence that various pre-analytical factors influence the mathematical interpretation of the data obtained. With an aim to contribute to the standardization and broaden the use of FC for human erythrocyte shape assessment, freshly prepared peripheral blood erythrocytes isolated from healthy donors were incubated in iso and hypo-osmotic solutions (pure saline, saline with potassium and calcium, and phosphate buffered saline) and examined by FC using values of forward scatter (FSC) and side scatter (SSC). Kurtosis, skewness, Pearson's second skewness coefficient of dissymmetry (PCD), and spherical index, calculated from FSC distributions, were used for the erythrocyte shape evaluation. In all isotonic media FSC distribution and FSC-based morphology parameters showed huge inter-individual and inter-medium variation. With decreasing osmolality, in all media and samples, the size of the erythrocytes increased, and swelling index and kurtosis decreased. However, changes in skewness and PCD were influenced by the medium used and the sample tested. Compared to FSC, SSC signal in isotonic and its change in hypotonic media showed lower inter-individual variation and was not influenced by the type of medium. We propose a spherical index and kurtosis as FSC-based indicators of erythrocyte shape. As more resistant to the influence of the preanalytical treatment, SSC data appeared to be unfairly neglected for the assessment of erythrocyte shape, in comparison to the usually employed FSC data.


Subject(s)
Erythrocytes , Flow Cytometry , Humans , Osmolar Concentration , Osmotic Fragility
8.
Cancer Immunol Immunother ; 68(4): 645-660, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30003321

ABSTRACT

Mounting evidence has accumulated on the critical role of the different myeloid cells in the regulation of the cancerous process, and in particular in the modulation of the immune reaction to cancer. Myeloid cells are a major component of host cells infiltrating tumors, interacting with each other, with tumor cells and other stromal cells, and demonstrating a prominent plasticity. We describe here various myeloid regulatory cells (MRCs) in mice and human as well as their relevant therapeutic targets. We first address the role of the monocytes and macrophages that can contribute to angiogenesis, immunosuppression and metastatic dissemination. Next, we discuss the differential role of neutrophil subsets in tumor development, enhancing the dual and sometimes contradicting role of these cells. A heterogeneous population of immature myeloid cells, MDSCs, was shown to be generated and accumulated during tumor progression as well as to be an important player in cancer-related immune suppression. Lastly, we discuss the role of myeloid DCs, which can either contribute to effective anti-tumor responses or play a more regulatory role. We believe that MRCs play a critical role in cancer-related immune regulation and suggest that future anti-cancer therapies will focus on these abundant cells.


Subject(s)
Cell Communication/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Animals , Biomarkers , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/pathology , Neutrophils/immunology , Neutrophils/metabolism
9.
J Cell Physiol ; 233(1): 447-462, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28295277

ABSTRACT

Lipopolysaccharide (LPS) is a pertinent deleterious factor in oral microenvironment for cells which are carriers of regenerative processes. The aim of this study was to investigate the emerging in vitro effects of LPS (Escherichia coli) on human periodontal ligament stem cell (PDLSC) functions and associated signaling pathways. We demonstrated that LPS did not affect immunophenotype, proliferation, viability, and cell cycle of PDLSCs. However, LPS modified lineage commitment of PDLSCs inhibiting osteogenesis by downregulating Runx2, ALP, and Ocn mRNA expression, while stimulating chondrogenesis and adipogenesis by upregulating Sox9 and PPARγ mRNA expression. LPS promoted myofibroblast-like phenotype of PDLSCs, since it significantly enhanced PDLSC contractility, as well as protein and/or gene expression of TGF-ß, fibronectin (FN), α-SMA, and NG2. LPS also increased protein and gene expression levels of anti-inflammatory COX-2 and pro-inflammatory IL-6 molecules in PDLSCs. Inhibition of peripheral blood mononuclear cells (MNCs) transendothelial migration in presence of LPS-treated PDLSCs was accompanied by the reduction of CD29 expression within MNCs. However, LPS treatment did not change the inhibitory effect of PDLSCs on mitogen-stimulated proliferation of CD4+ and the ratio of CD4+ CD25high /CD4+ CD25low lymphocytes. LPS-treated PDLSCs did not change the frequency of CD34+ and CD45+ cells, but decreased the frequency of CD33+ and CD14+ myeloid cells within MNCs. Moreover, LPS treatment attenuated the stimulatory effect of PDLSCs on CFC activity of MNCs, predominantly the CFU-GM number. The results indicated that LPS-activated ERK1,2 was at least partly involved in the observed effects on PDLSC differentiation capacity, acquisition of myofibroblastic attributes, and changes of their immunomodulatory features.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myofibroblasts/drug effects , Periodontal Ligament/drug effects , Stem Cells/drug effects , Adipogenesis/drug effects , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cellular Microenvironment , Chondrogenesis/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Myofibroblasts/enzymology , Myofibroblasts/immunology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/drug effects , PPAR gamma/genetics , PPAR gamma/metabolism , Periodontal Ligament/enzymology , Periodontal Ligament/immunology , Phenotype , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction/drug effects , Stem Cells/enzymology , Stem Cells/immunology , Time Factors , Transendothelial and Transepithelial Migration/drug effects
10.
Biochim Biophys Acta ; 1853(2): 431-44, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25433194

ABSTRACT

Mesenchymal stem cells (MSCs) have the potential to migrate toward damaged tissues increasing tissue regeneration. Interleukin-17 (IL-17) is a proinflammatory cytokine with pleiotropic effects associated with many inflammatory diseases. Although IL-17 can modulate MSC functions, its capacity to regulate MSC migration is not well elucidated so far. Here, we studied the role of IL-17 on peripheral blood (PB) derived MSC migration and transmigration across endothelial cells. IL-17 increased PB-MSC migration in a wound healing assay as well as cell mobilization from collagen gel. Concomitantly IL-17 induced the expression of urokinase type plasminogen activator (uPA) without affecting matrix metalloproteinase expression. The incremented uPA expression mediated the capacity of IL-17 to enhance PB-MSC migration in a ERK1,2 MAPK dependent way. Also, IL-17 induced PB-MSC migration alongside with changes in cell polarization and uPA localization in cell protrusions. Moreover, IL-17 increased PB-MSC adhesion to endothelial cells and transendothelial migration, as well as increased the capacity of PB-MSC adhesion to fibronectin, in an uPA-dependent fashion. Therefore, our data suggested that IL-17 may act as chemotropic factor for PB-MSCs by incrementing cell motility and uPA expression during inflammation development.


Subject(s)
Blood Cells/cytology , Cell Movement/drug effects , Interleukin-17/pharmacology , Mesenchymal Stem Cells/cytology , Transendothelial and Transepithelial Migration/drug effects , Urokinase-Type Plasminogen Activator/metabolism , Animals , Blood Cells/drug effects , Blood Cells/enzymology , Cell Adhesion/drug effects , Cell Line , Cell Polarity/drug effects , Collagen/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/metabolism , Humans , Immunophenotyping , Matrix Metalloproteinases/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Mice , Receptors, Interleukin-17/metabolism
11.
Histochem Cell Biol ; 146(3): 311-24, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27129368

ABSTRACT

Macrophage migration inhibitory factor is a well-known proinflammatory cytokine that is released during systemic stress response. Although MIF can affect erythrocyte production, the role of this cytokine in stress-induced erythropoiesis is completely unknown. To extend our previous findings showing that chronic psychological stress stimulates extramedullary erythropoiesis, here we examined whether MIF is involved in the control of stress-induced erythropoietic response. Adult male C57BL/6 wild-type (WT) and MIF-KO (knock-out) mice were subjected to 2-h daily restraint stress for either 7 or 14 consecutive days. The number of erythroid progenitors and CD71/Ter119 profile of erythroid precursors were analyzed in the bone marrow and spleen. Additionally, MIF protein expression was assessed in WT mice. Our results demonstrated that chronic restraint stress enhanced the number of both erythroid progenitors and precursors in the spleen. Stress-induced increase in the number of splenic late erythroid progenitors as well as in the percentage of CD71(+)Ter119(+)-double-positive precursors was significantly more pronounced in MIF-KO mice compared to WT animals. Furthermore, repeatedly stressed WT animals demonstrated an augmented MIF expression in the spleen. Unlike the spleen, the bone marrow of chronically stressed WT mice exhibited less prominent changes in erythropoietic stress response and no significant alteration in MIF expression. In addition, MIF deficiency did not influence the bone marrow erythropoiesis in stressed animals. These findings suggest that MIF regulates extramedullary erythropoiesis by inhibiting an overexpansion of splenic immature erythroid cells during chronic stress and indicate a novel role for this cytokine under chronic stress conditions.


Subject(s)
Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythropoiesis , Macrophage Migration-Inhibitory Factors/metabolism , Stress, Physiological , Animals , Macrophage Migration-Inhibitory Factors/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
IUBMB Life ; 68(3): 190-200, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26805406

ABSTRACT

Mesenchymal stem cells from human adipose tissue (hASCs) are proposed as suitable tools for soft tissue engineering and reconstruction. Although it is known that hASCs have the ability to home to sites of inflammation and tumor niche, the role of inflammatory cytokines in the hASCs-affected tumor development is not understood. We found that interferon-γ (IFN-γ) and/or tumor necrosis factor-α (TNF-α) prime hASCs to produce soluble factors which enhance MCF-7 cell line malignancy in vitro. IFN-γ and/or TNF-α-primed hASCs produced conditioned media (CM) which induced epithelial to mesenchymal transition (EMT) of MCF-7 cells by reducing E-Cadherin and increasing Vimentin expression. Induced EMT was accompanied by increased invasion, migration, and urokinase type-plasminogen activator (uPA) expression in MCF-7 cells. These effects were mediated by increased expression of transforming growth factor-ß1(TGF-ß1) in cytokines-primed hASCs, since inhibition of type I TGF-ß1 receptor on MCF-7 cells and neutralization of TGF-ß1 disabled the CM from primed hASCs to increase EMT, cell migration, and uPA expression in MCF-7 cells. Obtained data suggested that IFN-γ and/or TNF-α primed hASCs might enhance the malignancy of MCF-7 cell line by inducing EMT, cell motility and uPA expression in these cells via TGF-ß1-Smad3 signalization, with potentially important implications in breast cancer progression.


Subject(s)
Mesenchymal Stem Cells/physiology , Transforming Growth Factor beta1/physiology , Adipose Tissue/pathology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Epithelial-Mesenchymal Transition , Female , Humans , Interferon-gamma/physiology , MCF-7 Cells , Neoplasm Invasiveness , Signal Transduction , Tumor Necrosis Factor-alpha/physiology , Urokinase-Type Plasminogen Activator/physiology
13.
Mediators Inflamm ; 2016: 7314016, 2016.
Article in English | MEDLINE | ID: mdl-27630452

ABSTRACT

State of tumor microenvironment (TME) is closely linked to regulation of tumor growth and progression affecting the final outcome, refractoriness, and relapse of disease. Interactions of tumor, immune, and mesenchymal stromal/stem cells (MSCs) have been recognized as crucial for understanding tumorigenesis. Due to their outstanding features, stem cell-like properties, capacity to regulate immune response, and dynamic functional phenotype dependent on microenvironmental stimuli, MSCs have been perceived as important players in TME. Signals provided by tumor-associated chronic inflammation educate MSCs to alter their phenotype and immunomodulatory potential in favor of tumor-biased state of MSCs. Adjustment of phenotype to TME and acquisition of tumor-promoting ability by MSCs help tumor cells in maintenance of permissive TME and suppression of antitumor immune response. Potential utilization of MSCs in treatment of tumor is based on their inherent ability to home tumor tissue that makes them suitable delivery vehicles for immune-stimulating factors and vectors for targeted antitumor therapy. Here, we review data regarding intrusive effects of inflammatory TME on MSCs capacity to affect tumor development through modification of their phenotype and interactions with immune system.


Subject(s)
Mesenchymal Stem Cells/pathology , Neoplasms/pathology , Animals , Carcinogenesis/immunology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Humans , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Tumor Microenvironment/physiology
14.
Mediators Inflamm ; 2016: 2939658, 2016.
Article in English | MEDLINE | ID: mdl-28042204

ABSTRACT

Interleukin 17 (IL-17) is a cytokine with pleiotropic effects associated with several inflammatory diseases. Although elevated levels of IL-17 have been described in inflammatory myopathies, its role in muscle remodeling and regeneration is still unknown. Excessive extracellular matrix degradation in skeletal muscle is an important pathological consequence of many diseases involving muscle wasting. In this study, the role of IL-17 on the expression of matrix metalloproteinase- (MMP-) 9 in myoblast cells was investigated. The expression of MMP-9 after IL-17 treatment was analyzed in mouse myoblasts C2C12 cell line. The increase in MMP-9 production by IL-17 was concomitant with its capacity to inhibit myogenic differentiation of C2C12 cells. Doxycycline (Doxy) treatment protected the myogenic capacity of myoblasts from IL-17 inhibition and, moreover, increased myotubes hypertrophy. Doxy blocked the capacity of IL-17 to stimulate MMP-9 production by regulating IL-17-induced ERK1/2 MAPK activation. Our results imply that MMP-9 mediates IL-17's capacity to inhibit myoblast differentiation during inflammatory diseases and indicate that Doxy can modulate myoblast response to inflammatory induction by IL-17.


Subject(s)
Doxycycline/chemistry , Interleukin-17/metabolism , MAP Kinase Signaling System , Matrix Metalloproteinase 9/metabolism , Muscle Development , Myoblasts/cytology , Animals , Cell Differentiation , Cell Line , Gene Expression Regulation , Inflammation , Mice , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Recombinant Proteins/metabolism
15.
Blood Cells Mol Dis ; 55(4): 373-81, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26460262

ABSTRACT

PURPOSE: We compared the gene expression profile of peripheral blood CD34(+) cells and granulocytes in subjects with chronic myeloid leukemia (CML), with the accent on signaling pathways affected by BCR-ABL oncogene. METHODS: The microarray analyses have been performed in circulating CD34(+) cells and granulocytes from peripheral blood of 7 subjects with CML and 7 healthy donors. All studied BCR-ABL positive CML patients were in chronic phase, with a mean value of 2012±SD of CD34(+)cells/µl in peripheral blood. RESULTS: The gene expression profile was more prominent in CML CD34(+) cells (3553 genes) compared to granulocytes (2701 genes). The 41 and 39 genes were significantly upregulated in CML CD34(+) cells (HINT1, TXN, SERBP1) and granulocytes, respectively. BCR-ABL oncogene activated PI3K/AKT and MAPK signaling through significant upregulation of PTPN11, CDK4/6, and MYC and reduction of E2F1, KRAS, and NFKBIA gene expression in CD34(+) cells. Among genes linked to the inhibition of cellular proliferation by BCR-ABL inhibitor Imatinib, the FOS and STAT1 demonstrated significantly decreased expression in CML. CONCLUSION: The presence of BCR-ABL fusion gene doubled the expression quantity of genes involved in the regulation of cell cycle, proliferation and apoptosis of CD34(+) cells. These results determined the modified genes in PI3K/AKT and MAPK signaling of CML subjects.


Subject(s)
Antigens, CD34/metabolism , Gene Expression Regulation, Leukemic , Granulocytes/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Neoplastic Stem Cells/metabolism , Transcriptome , Biomarkers , Case-Control Studies , Cluster Analysis , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Profiling , Granulocytes/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Signal Transduction
16.
Mediators Inflamm ; 2015: 470458, 2015.
Article in English | MEDLINE | ID: mdl-25999667

ABSTRACT

Adult stem cells have a great potential applicability in regenerative medicine and cell-based therapies. However, there are still many unresolved issues concerning their biology, and the influence of the local microenvironment on properties of stem cells has been increasingly recognized. Interleukin (IL-) 17, as a cytokine implicated in many physiological and pathological processes, should be taken into consideration as a part of a regulatory network governing tissue-associated stem cells' fate. This review is focusing on the published data on the effects of IL-17 on the properties and function of hematopoietic and mesenchymal stem cells and trying to discuss that IL-17 achieves many of its roles by acting on adult stem cells.


Subject(s)
Hematopoietic Stem Cells/metabolism , Interleukin-17/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Humans
17.
J Cell Mol Med ; 18(1): 91-103, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24283209

ABSTRACT

Psychological stress affects different physiological processes including haematopoiesis. However, erythropoietic effects of chronic psychological stress remain largely unknown. The adult spleen contains a distinct microenvironment favourable for rapid expansion of erythroid progenitors in response to stressful stimuli, and emerging evidence suggests that inappropriate activation of stress erythropoiesis may predispose to leukaemic transformation. We used a mouse model to study the influence of chronic psychological stress on erythropoiesis in the spleen and to investigate potential mediators of observed effects. Adult mice were subjected to 2 hrs daily restraint stress for 7 or 14 consecutive days. Our results showed that chronic exposure to restraint stress decreased the concentration of haemoglobin in the blood, elevated circulating levels of erythropoietin and corticosterone, and resulted in markedly increased number of erythroid progenitors and precursors in the spleen. Western blot analysis revealed significantly decreased expression of both erythropoietin receptor and glucocorticoid receptor in the spleen of restrained mice. Furthermore, chronic stress enhanced the expression of stem cell factor receptor in the red pulp. Moreover, chronically stressed animals exhibited significantly increased expression of bone morphogenetic protein 4 (BMP4) in the red pulp as well as substantially enhanced mRNA expression levels of its receptors in the spleen. These findings demonstrate for the first time that chronic psychological stress activates BMP4-dependent extramedullary erythropoiesis and leads to the prolonged activation of stress erythropoiesis pathways. Prolonged activation of these pathways along with an excessive production of immature erythroid cells may predispose chronically stressed subjects to a higher risk of leukaemic transformation.


Subject(s)
Bone Morphogenetic Protein 4/physiology , Erythropoiesis , Stress, Psychological/physiopathology , Animals , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Chronic Disease , Corticosterone/blood , Down-Regulation , Erythropoietin/blood , Hemoglobins/metabolism , Iron/blood , Male , Mice , Mice, Inbred CBA , Organ Size , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Erythropoietin/metabolism , Receptors, Glucocorticoid/metabolism , Restraint, Physical , Signal Transduction , Spleen/pathology , Spleen/physiopathology , Stress, Psychological/blood
18.
Cell Biol Int ; 38(2): 254-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24155046

ABSTRACT

Adipose tissue is an attractive source of mesenchymal stem/stromal cells (MSCs) with potential applications in reconstructive plastic surgery and regenerative medicine. The aim of this study was to characterise human adipose tissue MSCs (ASCs) derived from healthy individuals and cancer patients and to compare their interactions with tumour cells. ASCs were isolated from adipose tissue of healthy donors, breast cancer-adjacent adipose tissue of breast cancer patients and tumour-adjacent adipose tissue of non-breast cancer patients. Their proliferation, differentiation, immunophenotype and gene expression were assessed and effects on the proliferation of human breast cancer cell line MCF-7 compared. ASCs from all sources exhibited similar morphology, proliferative and differentiation potential, showing the characteristic pattern of mesenchymal surface markers expression (CD90, CD105, CD44H, CD73) and the lack of HLA-DR and hematopoietic markers (CD11a, CD33, CD45, Glycophorin-CD235a), but uneven expression of CD34. ASCs also shared a common positive gene expression of HLA-DR, HLA-A, IL-6, TGF-ß and HIF-1, but were negative for HLA-G, while the expression levels of Cox-2 and IDO-1 varied. All ASCs significantly stimulated the proliferation of MCF-7 tumour cells in direct mixed co-cultures and transwell system, although their conditioned media displayed antiproliferative activity. Data obtained showed that ASCs with similar characteristics are easily isolated from various donors and sites of origin, although ASCs could both suppress and favour tumour cells growth, emphasising the importance of cellular context within the microenvironment and pointing to the significance of safety studies to exclude any potential clinical risk of their application in regenerative medicine.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/pathology , Breast Neoplasms/pathology , MCF-7 Cells/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/pathology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Breast/immunology , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , MCF-7 Cells/cytology , MCF-7 Cells/immunology , MCF-7 Cells/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism
19.
Transl Oncol ; 40: 101871, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38134841

ABSTRACT

Among multiple hemostasis components, platelets hyperactivity plays major roles in cancer progression by providing surface and internal components for intercellular crosstalk as well as by behaving like immune cells. Since platelets participate and regulate immunity in homeostatic and disease states, we assumed that revealing platelets profile might help in conceiving novel anti-cancer immune-based strategies. The goal of this review is to compile and discuss the most recent reports on the nature of cancer-associated platelets and their interference with immunotherapy. An increasing number of studies have emphasized active communication between cancer cells and platelets, with platelets promoting cancer cell survival, growth, and metastasis. The anti-cancer potential of platelet-directed therapy has been intensively investigated, and anti-platelet agents may prevent cancer progression and improve the survival of cancer patients. Platelets can (i) reduce antitumor activity; (ii) support immunoregulatory cells and factors generation; (iii) underpin metastasis and, (iv) interfere with immunotherapy by expressing ligands of immune checkpoint receptors. Mediators produced by tumor cell-induced platelet activation support vein thrombosis, constrain anti-tumor T- and natural killer cell response, while contributing to extravasation of tumor cells, metastatic potential, and neovascularization within the tumor. Recent studies showed that attenuation of immunothrombosis, modulation of platelets and their factors have a good perspective in immunotherapy optimization. Particularly, blockade of intra-tumoral platelet-associated programmed death-ligand 1 might promote anti-tumor T cell-induced cytotoxicity. Collectively, these findings suggest that platelets might represent the source of relevant cancer staging biomarkers, as well as promising targets and carriers in immunotherapeutic approaches for combating cancer.

20.
Biochim Biophys Acta ; 1823(4): 838-49, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22285818

ABSTRACT

The present study evaluated the role of interleukin (IL) 17 in multilineage commitment of C2C12 myoblastic cells and investigated associated signaling pathways. The results concerning the effects on cell function showed that IL-17 inhibits the migration of C2C12 cells, while not affecting their proliferation. The data regarding the influence on differentiation demonstrated that IL-17 inhibits myogenic differentiation of C2C12 cells by down-regulating the myogenin mRNA level, myosin heavy chain expression and myotube formation, but promotes their osteogenic differentiation by up-regulating the Runt-related transcription factor 2 mRNA level, cyclooxygenase-2 expression and alkaline phosphatase activity. IL-17 exerted these effects by activating ERK1,2 mitogen activated protein kinase signaling pathway, which in turn regulated the expression of relevant genes and proteins to inhibit myogenic differentiation and induce osteogenic differentiation. Additional analysis showed that the induction of osteogenic differentiation by IL-17 is independent of BMP signaling. The results obtained demonstrate the potential of IL-17 not only to inhibit the myogenic differentiation of C2C12 myoblasts but also to convert their differentiation pathway into that of osteoblast lineage providing new insight into the capacities of IL-17 to modulate the differentiation commitment.


Subject(s)
Cell Differentiation/drug effects , Interleukin-17/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Development/drug effects , Myoblasts/cytology , Osteogenesis/drug effects , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , MAP Kinase Signaling System/drug effects , Mice , Models, Biological , Myoblasts/drug effects , Myoblasts/enzymology , Receptors, Interleukin-17/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL