Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Wound Care ; 32(7): 422-427, 2023 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-37405941

RESUMEN

OBJECTIVE: By default, the antimicrobial efficacy of antiseptics used in wound management is tested in vitro under standardised conditions according to European standard DIN EN 13727, with albumin and sheep erythrocytes used as organic challenge. However, it is not clear whether these testing conditions adequately reflect the wound bed environment and its interaction with antiseptic products intended to be used in wounds in humans. METHOD: This study compared the efficacy of different commercial antiseptic products based on octenidine dihydrochloride (OCT), polyhexamethylene biguanide (PHMB) and povidone-iodine under challenge with human wound exudate collected from the hard-to-heal wounds of patients, compared to the standardised organic load, in an in vitro setting according to DIN EN 13727. RESULTS: The bactericidal efficacy of the tested products was reduced to a different extent when challenged with human wound exudate, compared to the standardised conditions. Overall, OCT-based products showed the necessary germ count reductions at the shortest exposure times (e.g., 15 seconds for Octenisept (Schülke & Mayr GmbH, Germany)). PHMB-based products were the least efficient. In addition to the protein content, other components of wound exudate, such as the microbiota, seem to influence the efficacy of antiseptics. CONCLUSION: This study demonstrated that the standardised in vitro test conditions may only partially reflect actual wound bed conditions in humans.


Asunto(s)
Antiinfecciosos Locales , Antiinfecciosos , Humanos , Antibacterianos/farmacología , Antiinfecciosos Locales/farmacología , Exudados y Transudados , Povidona Yodada/farmacología
2.
Stem Cells ; 39(9): 1270-1284, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34013984

RESUMEN

Acute myeloid leukemia (AML) is characterized by an expansion of leukemic cells and a simultaneous reduction of normal hematopoietic precursors in the bone marrow (BM) resulting in hematopoietic insufficiency, but the underlying mechanisms are poorly understood in humans. Assuming that leukemic cells functionally inhibit healthy CD34+ hematopoietic stem and progenitor cells (HSPC) via humoral factors, we exposed healthy BM-derived CD34+ HSPC to cell-free supernatants derived from AML cell lines as well as from 24 newly diagnosed AML patients. Exposure to AML-derived supernatants significantly inhibited proliferation, cell cycling, colony formation, and differentiation of healthy CD34+ HSPC. RNA sequencing of healthy CD34+ HSPC after exposure to leukemic conditions revealed a specific signature of genes related to proliferation, cell-cycle regulation, and differentiation, thereby reflecting their functional inhibition on a molecular level. Experiments with paired patient samples showed that these inhibitory effects are markedly related to the immunomagnetically enriched CD34+ leukemic cell population. Using PCR, ELISA, and RNA sequencing, we detected overexpression of TGFß1 in leukemic cells on the transcriptional and protein level and, correspondingly, a molecular signature related to TGFß1 signaling in healthy CD34+ HSPC. This inhibitory effect of TGFß1 on healthy hematopoiesis was functionally corrobated and could be pharmacologically reverted by SD208, an inhibitor of TGFß receptor 1 signaling. Overall, these data indicate that leukemic cells induce functional inhibition of healthy CD34+ HSPC, at least in part, through TGFß1, suggesting that blockage of this pathway may improve hematopoiesis in AML.


Asunto(s)
Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Antígenos CD34/metabolismo , Médula Ósea/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética
3.
Diabetologia ; 64(8): 1834-1849, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34131781

RESUMEN

AIMS/HYPOTHESIS: People with diabetes have an increased cardiovascular risk with an accelerated development of atherosclerosis and an elevated mortality rate after myocardial infarction. Therefore, cardioprotective effects of glucose-lowering therapies are of major importance for the pharmacotherapy of individuals with type 2 diabetes. For sodium-glucose cotransporter 2 inhibitors (SGLT2is), in addition to a reduction in blood glucose, beneficial effects on atherosclerosis, obesity, renal function and blood pressure have been observed. Recent results showed a reduced risk of worsening heart failure and cardiovascular deaths under dapagliflozin treatment irrespective of the diabetic state. However, the underlying mechanisms are yet unknown. Platelets are known drivers of atherosclerosis and atherothrombosis and disturbed platelet activation has also been suggested to occur in type 2 diabetes. Therefore, the present study investigates the impact of the SGLT2i dapagliflozin on the interplay between platelets and inflammation in atherogenesis. METHODS: Male, 8-week-old LDL-receptor-deficient (Ldlr-/-) mice received a high-fat, high-sucrose diabetogenic diet supplemented without (control) or with dapagliflozin (5 mg/kg body weight per day) for two time periods: 8 and 25 weeks. In a first translational approach, eight healthy volunteers received 10 mg dapagliflozin/day for 4 weeks. RESULTS: Dapagliflozin treatment ameliorated atherosclerotic lesion development, reduced circulating platelet-leucocyte aggregates (glycoprotein [GP]Ib+CD45+: 29.40 ± 5.94 vs 17.00 ± 5.69 cells, p < 0.01; GPIb+lymphocyte antigen 6 complex, locus G+ (Ly6G): 8.00 ± 2.45 vs 4.33 ± 1.75 cells, p < 0.05) and decreased aortic macrophage infiltration (1.31 ± 0.62 vs 0.70 ± 0.58 ×103 cells/aorta, p < 0.01). Deeper analysis revealed that dapagliflozin decreased activated CD62P-positive platelets in Ldlr-/- mice fed a diabetogenic diet (3.78 ± 1.20% vs 2.83 ± 1.06%, p < 0.01) without affecting bleeding time (85.29 ± 37.27 vs 89.25 ± 16.26 s, p = 0.78). While blood glucose was only moderately affected, dapagliflozin further reduced endogenous thrombin generation (581.4 ± 194.6 nmol/l × min) × 10-9 thrombin vs 254.1 ± 106.4 (nmol/l × min) × 10-9 thrombin), thereby decreasing one of the most important platelet activators. We observed a direct inhibitory effect of dapagliflozin on isolated platelets. In addition, dapagliflozin increased HDL-cholesterol levels. Importantly, higher HDL-cholesterol levels (1.70 ± 0.58 vs 3.15 ± 1.67 mmol/l, p < 0.01) likely contribute to dapagliflozin-mediated inhibition of platelet activation and thrombin generation. Accordingly, in line with the results in mice, treatment with dapagliflozin lowered CD62P-positive platelet counts in humans after stimulation by collagen-related peptide (CRP; 88.13 ± 5.37% of platelets vs 77.59 ± 10.70%, p < 0.05) or thrombin receptor activator peptide-6 (TRAP-6; 44.23 ± 15.54% vs 28.96 ± 11.41%, p < 0.01) without affecting haemostasis. CONCLUSIONS/INTERPRETATION: We demonstrate that dapagliflozin-mediated atheroprotection in mice is driven by elevated HDL-cholesterol and ameliorated thrombin-platelet-mediated inflammation without interfering with haemostasis. This glucose-independent mechanism likely contributes to dapagliflozin's beneficial cardiovascular risk profile.


Asunto(s)
Compuestos de Bencidrilo/uso terapéutico , Enfermedad de la Arteria Coronaria/prevención & control , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucósidos/uso terapéutico , Activación Plaquetaria/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Trombina/metabolismo , Adulto , Animales , Glucemia/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control , HDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Citometría de Flujo , Voluntarios Sanos , Humanos , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Selectina-P/metabolismo , Recuento de Plaquetas , Reacción en Cadena en Tiempo Real de la Polimerasa , Conducta de Reducción del Riesgo
4.
J Biol Chem ; 291(8): 4091-106, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26699196

RESUMEN

The aim of this study was to characterize the interaction of KYSE-410, an esophageal squamous cell carcinoma cell line, and fibroblasts with respect to the extracellular matrix component hyaluronan (HA) and chemokine expression. KYSE-410 cells induced the mRNA expression of HA synthase 2 (Has2) in normal skin fibroblasts (SF) only in direct co-cultures. Parallel to Has2 mRNA, Has2 antisense RNA (Has2os2) was up-regulated in co-cultures. Knockdown of LEF1, a downstream target of Wnt signaling, abrogated Has2 and Has2os2 induction. After knockdown of Has2 in SF, significantly less α-smooth muscle actin expression was detected in co-cultures. Moreover, it was investigated whether the phenotype of KYSE-410 was affected in co-culture with SF and whether Has2 knockdown in SF had an impact on KYSE-410 cells in co-culture. However, no effects on epithelial-mesenchymal transition markers, proliferation, and migration were detected. In addition to Has2 mRNA, the chemokine CCL5 was up-regulated and CCL11 was down-regulated in SF in co-culture. Furthermore, co-cultures of KYSE-410 cells and cancer-associated fibroblasts (CAF) were investigated. Similar to SF, Has2 and Ccl5 were up-regulated and Ccl11 was down-regulated in CAF in co-culture. Importantly and in contrast to SF, inhibiting HA synthesis by 4-methylumbelliferone abrogated the effect of co-culture on Ccl5 in CAF. Moreover, HA was found to promote adhesion of CD4(+) but not CD8(+) cells to xenogaft tumor tissues. In conclusion, direct co-culture of esophageal squamous cell carcinoma and fibroblasts induced stromal HA synthesis via Wnt/LEF1 and altered the chemokine profile of stromal fibroblasts, which in turn may affect the tumor immune response.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Quimiocina CCL11/biosíntesis , Quimiocina CCL5/biosíntesis , Neoplasias Esofágicas/metabolismo , Fibroblastos/metabolismo , Ácido Hialurónico/biosíntesis , Proteínas de Neoplasias/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Quimiocina CCL11/genética , Quimiocina CCL5/genética , Técnicas de Cocultivo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Fibroblastos/patología , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Humanos , Hialuronano Sintasas , Ácido Hialurónico/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Vía de Señalización Wnt
5.
Int J Cancer ; 141(4): 791-804, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28493326

RESUMEN

Epidemiological studies have detected a higher incidence of various tumour entities in diabetic patients. However, the underlying mechanisms remain insufficiently understood. Glucose-derived pericellular and extracellular hyaluronan (HA) promotes tumour progression and development. In our study, we tested the hypothesis that a diabetic metabolic state, characterised by hyperglycaemia and concomitant aberrant insulin signalling, stimulates tumour progression via the induction of HA synthesis. In a streptozotocin-induced diabetic nude mouse tumour xenograft model, hyperglycaemia and lack of insulin caused an increased formation of tumour-associated HA-matrix, which in turn accelerated tumour progression and neoangiogenesis. This process was effectively attenuated by treatment with 4-methylumbelliferone, a pharmacological inhibitor of HA-synthesis. To define the mechanisms behind these in vivo observations, we investigated the impact of hyperglycaemia and insulin on the glucose metabolism in oesophageal squamous cell cancer cells (ESCC). Hyperglycaemia induced HA synthesis while insulin diminished HA production by directing glucose metabolites to glycolysis. Vice versa, inhibition of glycolysis, either by knockdown of the glycolytic key enzyme phosphofructokinase or by an experimental abrogation of insulin signalling (knockdown of the insulin receptor and long-term treatment with insulin) augmented HA synthesis. Consequently, these processes induced invasion, anchorage-independent growth and adhesion of ESCC to endothelial cells in vitro. Thus, the cellular shift in glucose usage from catabolism of glucose to anabolism of HA driven by hyperglycaemia and insulin resistance may represent an important link between diabetes and cancer progression. Hence, therapeutical inhibition of HA synthesis may represent a promising approach for tumour treatment in diabetic patients.


Asunto(s)
Carcinoma de Células Escamosas/patología , Diabetes Mellitus Experimental/metabolismo , Neoplasias Esofágicas/patología , Ácido Hialurónico/metabolismo , Hiperglucemia/complicaciones , Insulina/metabolismo , Animales , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Diabetes Mellitus Experimental/complicaciones , Progresión de la Enfermedad , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Transducción de Señal
6.
Arterioscler Thromb Vasc Biol ; 36(2): e9-16, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26586662

RESUMEN

OBJECTIVE: Hyaluronan (HA) is a polymeric glucosaminoglycan that forms a provisional extracellular matrix in diseased vessels. HA is synthesized by 3 different HA synthases (HAS1, HAS2, and HAS3). Aim of this study was to unravel the role of the HAS3 isoenzyme during experimental neointimal hyperplasia. APPROACH AND RESULTS: Neointimal hyperplasia was induced in Has3-deficient mice by ligation of the carotid artery. HA in the media of Has3-deficient mice was decreased 28 days after ligation, and neointimal hyperplasia was strongly inhibited. However, medial and luminal areas were unaffected. Cell density, proliferation, and apoptosis were not altered, suggesting a proportional decrease of both, the number of cells and extracellular matrix. In addition, endothelial function as determined by acetylcholine-induced relaxation of aortic rings, immunoblotting of endothelial nitric oxide synthase, and arterial blood pressure were not affected. Furthermore, the oxidative stress response was not affected as determined in total protein extracts from aortae. Transcriptome analysis comparing control versus ligated carotid arteries hinted toward a mitigated differential regulation of various signaling pathways in Has3-deficient mice in response to ligation that were related to vascular smooth muscle cell (VSMC) migration, including focal adhesions, integrins, mitogen-activated protein kinase, and phosphatidylinositol signaling system. Lentiviral overexpression of HAS3 in VSMC supported the migratory phenotype of VSMC in response to platelet-derived growth factor BB in vitro. Accordingly, knockdown of HAS3 reduced the migratory response to platelet-derived growth factor BB and in addition decreased the expression of PDGF-B mRNA. CONCLUSIONS: HAS3-mediated HA synthesis after vessel injury supports seminal signaling pathways in activation of VSMC, increases platelet-derived growth factor BB-mediated migration, and in turn enhances neointimal hyperplasia in vivo.


Asunto(s)
Enfermedades de las Arterias Carótidas/enzimología , Glucuronosiltransferasa/deficiencia , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Neointima , Animales , Becaplermina , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Movimiento Celular , Proliferación Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Genotipo , Glucuronosiltransferasa/genética , Hialuronano Sintasas , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fenotipo , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Transducción de Señal , Transcripción Genética , Transfección
7.
J Biol Chem ; 285(30): 23276-84, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20463012

RESUMEN

Hyaluronan (HA) is a polysaccharide component in the parenchyma and stroma of human esophageal squamous cell carcinoma (ESCC). Clinically, esophageal cancer represents a highly aggressive tumor type with poor prognosis resulting in a 5-year survival rate of 5%. The aim of the present study was the detailed analysis of the role of HA synthesis for ESCC phenotype in vitro using the ESCC cell line OSC1. In OSC1 cells, pericellular HA-matrix surrounding extended actin-dependent filopodia was detected. The small molecule inhibitor of HA synthesis, 4-methylumbelliferone (4-MU, 0.3 mm) caused loss of these filopodia and focal adhesions and inhibited proliferation and migration. In search of the underlying mechanism cleavage of focal adhesion kinase (FAK) was detected by immunoblotting. In addition, displacing HA by an HA-binding peptide (Pep-1, 500 mug/ml) and digestion of pericellular HA by hyaluronidase resulted in cleavage of focal adhesions. Furthermore, real-time reverse transcription PCR revealed that HA synthase 3 (HAS3) > HAS2 are the predominant HA-synthases in OSC1. Lentiviral transduction with shHAS3, and to a lesser extent with shHAS2, reduced intact FAK protein and filopodia as well as proliferation and migration. Furthermore, down-regulation by lentiviral shRNA of RHAMM (receptor of HA-mediated motility) but not CD44 induced loss of filopodia and caused FAK cleavage. In contrast, knockdown of both HA receptors inhibited proliferation and migration of OSC1. In conclusion, HA synthesis and, in turn, RHAMM and CD44 signaling promoted an activated phenotype of OSC1. Because RHAMM appears to support both filopodia, FAK, and the proliferative and migratory phenotype, it may be promising to explore RHAMM as a potential therapeutic target in esophageal cancer.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Adhesiones Focales/metabolismo , Ácido Hialurónico/metabolismo , Fenotipo , Seudópodos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Esofágicas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Adhesiones Focales/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glucuronosiltransferasa/deficiencia , Glucuronosiltransferasa/genética , Humanos , Receptores de Hialuranos/metabolismo , Hialuronano Sintasas , Ácido Hialurónico/biosíntesis , Himecromona/análogos & derivados , Himecromona/farmacología , Datos de Secuencia Molecular , Seudópodos/efectos de los fármacos
8.
Mol Cancer ; 10: 30, 2011 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-21429221

RESUMEN

BACKGROUND: Oesophageal cancer is a highly aggressive tumour entity with at present poor prognosis. Therefore, novel treatment options are urgently needed. Hyaluronan (HA) is a polysaccharide present in the matrix of human oesophageal squamous cell carcinoma (ESCC). Importantly, in vitro ESCC cells critically depend on HA synthesis to maintain the proliferative phenotype. The aim of the present study is (1) to study HA-synthase (HAS) expression and regulation in human ESCC, and (2) to translate the in vitro results into a mouse xenograft model of human ESCC to study the effects of systemic versus tumour targeted HAS inhibition on proliferation and distribution of tumour-bound and stromal hyaluronan. METHODS: mRNA expression was investigated in human ESCC biopsies by semiquantitative real-time RT PCR. Furthermore, human ESCC were xenografted into NMRI nu/nu mice. The effects on tumour progression and morphology of 4-methylumbelliferone (4-MU), an inhibitor of HA-synthesis, and of lentiviral knock down of HA-synthase 3 (HAS3), the main HAS isoform in the human ESCC tissues and the human ESCC cell line used in this study, were determined. Tumour progression was monitored by calliper measurements and by flat-panel detector volume computed tomography (fpVCT). HA content, cellular composition and proliferation (Ki67) were determined histologically. RESULTS: mRNA of HAS isoform 3 (HAS3) was upregulated in human ESCC biopsies and HAS3 mRNA was positively correlated to expression of the epidermal growth factor (EGF) receptor. EGF was also proven to be a strong inductor of HAS3 mRNA expression in vitro. During the course of seven weeks, 4-MU inhibited progression of xenograft tumours. Interestingly, remodelling of the tumour into a more differentiated phenotype and inhibition of cell proliferation were observed. Lentiviral knockdown of HAS3 in human ESCC cells prior to xenografting mimicked all effects of 4-MU treatment suggesting that hyaluronan produced by ESCC is accountable for major changes in tumour environment in vivo. CONCLUSIONS: Systemic inhibition of HA-synthesis and knockdown of tumour cell HAS3 cause decreased ESCC progression accompanied by tumour stroma remodelling and may therefore be used in novel approaches to ESCC therapy.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Glucuronosiltransferasa/metabolismo , Ácido Hialurónico/biosíntesis , Animales , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/prevención & control , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/prevención & control , Regulación Neoplásica de la Expresión Génica , Glucuronosiltransferasa/antagonistas & inhibidores , Glucuronosiltransferasa/genética , Humanos , Hialuronano Sintasas , Himecromona/análogos & derivados , Himecromona/farmacología , Inmunohistoquímica , Ratones , Ratones Endogámicos , Ratones Desnudos , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Curr Drug Targets ; 22(2): 202-213, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33001011

RESUMEN

With this article, we would like to take the reader on a journey into the world of molecular medicine as it has evolved over the past decades, enabled by advances in genomics. These findings advanced both the development of prognostic parameters and the evolvement of therapy strategies. In this manuscript, we will present haematopoietic diseases as a prime example of this progress because they are relevant not only for their frequency but also for the evident diagnostic and therapeutic progress. The growing understanding of the underlying pathophysiology originates from the cellular pathology as it was described by, e.g., Rudolf Virchow (1821-1902). The identification of specific genomic changes in haematological malignancies and solid tumour diseases provided us with very sensitive tools for diagnostics and prediction of prognosis. Thus, it paved the way for individualized or personalized therapy. In particular, the rapid development of sequencing techniques for the human genome using Next Generation Sequencing (NGS) has contributed to this progress. Recently, artificial intelligence provided us with the tools to analyze the complex interactions of genomic alterations, course of the disease, and further factors of as yet unknown significance. With all these indisputable improvements, we should not neglect the holistic treatment mandate of personalized therapy, i.e., therapy appropriate to the individual. In this context, the treating physician should address relevant co-morbidities, the psychosocial embedding of the patient and his desire for treatment.


Asunto(s)
Microscopía , Neoplasias , Medicina de Precisión , Inteligencia Artificial , Humanos , Leucemia Mieloide , Medicina de Precisión/tendencias , Pronóstico
10.
Sci Rep ; 11(1): 21229, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34707175

RESUMEN

Metastatic spread of cancer cells into a pre-metastatic niche is highly dependent on a supporting microenvironment. Human bone marrow-derived mesenchymal stem cells (bmMSCs) contribute to the tumor microenvironment and promote cancer metastasis by inducing epithelial-to-mesenchymal transition and immune evasion. The underlying mechanisms, however, are incompletely understood. The glycosaminoglycan hyaluronan (HA) is a central component of the extracellular matrix and has been shown to harbor pro-metastatic properties. In this study we investigated the highly disseminating breast cancer and glioblastoma multiforme cell lines MDA-MB-321 and U87-MG which strongly differ in their metastatic potential to evaluate the impact of HA on tumor promoting features of bmMSC and their interaction with tumor cells. We show that adipogenic differentiation of bmMSC is regulated by the HA-matrix. This study reveals that MDA-MB-231 cells inhibit this process by the induction of HA-synthesis in bmMSCs and thus preserve the pro-tumorigenic properties of bmMSC. Furthermore, we show that adhesion of MDA-MB-231 cells to bmMSC is facilitated by the tumor cell-induced HA-rich matrix and is mediated by the HA-receptor LAYN. We postulate that invasive breast cancer cells modulate the HA-matrix of bmMSC to adapt the pre-metastatic niche. Thus, the HA-matrix provides a potential novel therapeutic target to prevent cancer metastasis.


Asunto(s)
Diferenciación Celular , Ácido Hialurónico/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microambiente Tumoral , Adipocitos/citología , Adipocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Glioblastoma/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Células Madre Mesenquimatosas/citología
11.
Biomed Pharmacother ; 123: 109797, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31874445

RESUMEN

BACKGROUND: Non-allergic angioedema is largely driven by increased plasma levels of bradykinin and over-activation of bradykinin receptor type II (B2), but the specific downstream signalling pathways remain unclear. The aim of this study was to identify signal transduction events involved in bradykinin-induced dermal extravasation. METHODS: Quantification of dermal extravasation was accomplished following intradermal (i.d.) injection of bradykinin or the B2 agonist labradimil in mice with endothelial NO-synthase (eNOS) deficiency and in C57BL/6J mice pre-treated with vehicle, NO-synthase or cyclooxygenase (COX) inhibitors. In the multicentre clinical study ABRASE, 38 healthy volunteers received i.d. bradykinin injections into the ventral forearm before and after oral treatment with the COX inhibitor ibuprofen (600 mg). The primary endpoint of ABRASE was the mean time to complete resolution of wheals (TTCR) and the secondary endpoint was the change of maximal wheal size. RESULTS: Neither NOS inhibitors nor eNOS deficiency altered bradykinin-induced extravasation. In striking contrast, the COX inhibitors ibuprofen, diclofenac, SC560 and celecoxib significantly diminished this extravasation when given before injection. As for diclofenac, a similar but significantly lower effect was observed when given after i.d. injection of bradykinin. Similar results were obtained when bradykinin was replaced by labradimil. In volunteers, ibuprofen significantly reduced TTCR (P < 0.001) and maximal wheal size (P = 0.0044). CONCLUSION: These data suggest that COX activity contributes to bradykinin-induced dermal extravasation in mice and humans. In addition, our findings may open new treatment options and point to a potential activity of drugs interfering with the release of the COX substrate arachidonic acid, e.g. glucocorticoids.


Asunto(s)
Bradiquinina/farmacología , Dermis/patología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Inhibidores de la Ciclooxigenasa/farmacología , Extravasación de Materiales Terapéuticos y Diagnósticos , Humanos , Ratones Endogámicos C57BL
12.
Sci Rep ; 9(1): 1827, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755628

RESUMEN

Diabetic patients are at a greater risk of heart failure due to diabetic cardiomyopathy and worsened outcome post-myocardial infarction. While the molecular mechanisms remain unclear, fibrosis and chronic inflammation are common characteristics of both conditions. Diabetes mellitus (types I and II) results in excessive hyaluronan (HA) deposition in vivo, and hyperglycemia stimulates HA synthesis for several cell types in vitro. HA-rich extracellular matrix contributes to fibrotic, hyperplastic and inflammatory disease progression. We hypothesized that excessive hyperglycemia-driven HA accumulation may contribute to pathological fibroblast activation and fibrotic remodelling in diabetic patients. Therefore, we analysed the impact of both hyperglycemia and diet-induced obesity and insulin resistance on HA matrix formation and cardiac fibroblast activation. Here we report that cardiac fibroblasts isolated from mice on a diabetogenic diet acquire pro-fibrotic gene expression without a concomitant increase in HA matrix deposition. Additionally, hyperglycemia alone does not stimulate HA synthesis or cardiac fibroblast activation in vitro, suggesting that the direct effect of hyperglycemia on fibroblasts is not the primary driver of fibrotic remodelling in cardiac diabetic maladaptation.


Asunto(s)
Alimentación Animal , Fibroblastos/metabolismo , Ácido Hialurónico/metabolismo , Hiperglucemia/metabolismo , Resistencia a la Insulina , Miocardio/metabolismo , Animales , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Fibrosis , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Factor de Crecimiento Transformador beta1/metabolismo
13.
Br J Pharmacol ; 176(23): 4474-4490, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31351004

RESUMEN

BACKGROUND AND PURPOSE: Aerobic glycolysis is a unique feature of tumour cells that entails several advantages for cancer progression such as resistance to apoptosis. The low MW compound, dichloroacetate, is a pyruvate dehydrogenase kinase inhibitor, which restores oxidative phosphorylation and induces apoptosis in a variety of cancer entities. However, its therapeutic effectiveness is limited by resistance mechanisms. This study aimed to examine the role of the anti-apoptotic hyaluronan (HA) matrix in this context and to identify a potential add-on treatment option to overcome this limitation. EXPERIMENTAL APPROACH: The metabolic connection between dichloroacetate treatment and HA matrix augmentation was analysed in vitro by quantitative PCR and affinity cytochemistry. Metabolic pathways were analysed using Seahorse, HPLC, fluorophore-assisted carbohydrate electrophoresis, colourimetry, immunoblots, and immunochemistry. The effects of combining dichloroacetate with the HA synthesis inhibitor 4-methylumbelliferone was evaluated in 2D and 3D cell cultures and in a nude mouse tumour xenograft regression model by immunoblot, immunochemistry, and FACS analysis. KEY RESULTS: Mitochondrial reactivation induced by dichloroacetate metabolically activated HA synthesis by augmenting precursors as well as O-GlcNAcylation. This process was blocked by 4-methylumbelliferone, resulting in enhanced anti-tumour efficacy in 2D and 3D cell culture and in a nude mouse tumour xenograft regression model. CONCLUSIONS AND IMPLICATIONS: The HA rich tumour micro-environment represents a metabolic factor contributing to chemotherapy resistance. HA synthesis inhibition exhibited pronounced synergistic actions with dichloroacetate treatment on oesophageal tumour cell proliferation and survival in vitro and in vivo suggesting the combination of these two strategies is an effective anticancer therapy.


Asunto(s)
Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Matriz Extracelular/efectos de los fármacos , Ácido Hialurónico/antagonistas & inhibidores , Animales , Antineoplásicos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ácido Dicloroacético , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Matriz Extracelular/metabolismo , Humanos , Ácido Hialurónico/síntesis química , Ácido Hialurónico/metabolismo , Himecromona , Masculino , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Análisis de Regresión , Relación Estructura-Actividad , Células Tumorales Cultivadas
14.
Cancers (Basel) ; 11(11)2019 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-31671846

RESUMEN

Despite remarkable progress in melanoma therapy, the exceptional heterogeneity of the disease has prevented the development of reliable companion biomarkers for the prediction or monitoring of therapy responses. Here, we show that difficulties in detecting blood-based markers, like circulating tumor cells (CTC), might arise from the translation of the mutational heterogeneity of melanoma cells towards their surface marker expression. We provide a unique method, which enables the molecular characterization of clinically relevant CTC subsets, as well as circulating tumor DNA (ctDNA), from a single blood sample. The study demonstrates the benefit of a combined analysis of ctDNA and CTC counts in melanoma patients, revealing that CTC subsets and ctDNA provide synergistic real-time information on the mutational status, RNA and protein expression of melanoma cells in individual patients, in relation to clinical outcome.

15.
Oncotarget ; 7(16): 21428-40, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-26870892

RESUMEN

Biomarkers for prognosis in radiotherapy-treated breast cancer patients are urgently needed and important to stratify patients for adjuvant therapies. Recently, a role of the receptor of hyaluronan-mediated motility (RHAMM) has been suggested for tumor progression. Our aim was (i) to investigate the prognostic value of RHAMM in breast cancer and (ii) to unravel its potential function in the radiosusceptibility of breast cancer cells. We demonstrate that RHAMM mRNA expression in breast cancer biopsies is inversely correlated with tumor grade and overall survival. Radiosusceptibility in vitro was evaluated by sub-G1 analysis (apoptosis) and determination of the proliferation rate. The potential role of RHAMM was addressed by short interfering RNAs against RHAMM and its splice variants. High expression of RHAMMv1/v2 in p53 wild type cells (MCF-7) induced cellular apoptosis in response to ionizing radiation. In comparison, in p53 mutated cells (MDA-MB-231) RHAMMv1/v2 was expressed sparsely resulting in resistance towards irradiation induced apoptosis. Proliferation capacity was not altered by ionizing radiation in both cell lines. Importantly, pharmacological inhibition of the major ligand of RHAMM, hyaluronan, sensitized both cell lines towards radiation induced cell death. Based on the present data, we conclude that the detection of RHAMM splice variants in correlation with the p53 mutation status could help to predict the susceptibility of breast cancer cells to radiotherapy. Additionally, our studies raise the possibility that the response to radiotherapy in selected cohorts may be improved by pharmaceutical strategies against RHAMM and its ligand hyaluronan.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular/genética , Proteínas de la Matriz Extracelular/genética , Receptores de Hialuranos/genética , Empalme del ARN , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Células MCF-7 , Pronóstico , Interferencia de ARN , Tolerancia a Radiación/genética , Radiación Ionizante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia
17.
Am J Pathol ; 171(5): 1451-61, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17982124

RESUMEN

Remodeling of the dermal extracellular matrix occurs during photoaging. Here, the effect of repetitive UVB irradiation on dermal hyaluronic acid (HA) was examined. C57/BL6 mice were chronically (182 days) irradiated with UVB, and consecutive skin biopsies were collected during the irradiation period and afterward (300 and 400 days of age). UVB caused marked loss of HA from the papillary dermis and down-regulation of HA synthase 1 (HAS1), HAS2, and HAS3 mRNA expression. In contrast, hyaluronidases (HYAL) 1, HYAL2, and HA receptor CD44 were unchanged. Furthermore, transforming growth factor beta-1 (TGF-beta1) and TGF-beta1-receptor II expression were decreased in UVB-irradiated biopsies, and TGF-beta1 strongly induced HAS1 and HAS2 expression in cultured dermal fibroblasts. Therefore, TGF-beta1 might be one factor involved in UVB-induced down-regulation of HAS enzymes. In addition, total cell number and the percentage of proliferating fibroblasts in the papillary dermis of UVB-irradiated mice were decreased. Down-regulation of HAS2 by lentiviral overexpression of short hairpin RNA in vitro caused inhibition of HA synthesis, DNA synthesis, and migration of dermal fibroblasts. In conclusion, chronic UVB irradiation induces loss of HA from the dermis, thereby contributing to the quiescent phenotype of dermal fibroblasts.


Asunto(s)
Dermis/efectos de la radiación , Glucuronosiltransferasa/biosíntesis , Ácido Hialurónico/metabolismo , Rayos Ultravioleta/efectos adversos , Animales , Proliferación Celular , Células Cultivadas , Quimiotaxis , Dermis/citología , Dermis/metabolismo , Regulación hacia Abajo , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibroblastos/efectos de la radiación , Glucuronosiltransferasa/genética , Humanos , Receptores de Hialuranos/metabolismo , Hialuronano Sintasas , Hialuronoglucosaminidasa/metabolismo , Isoenzimas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA