Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Eur Rev Med Pharmacol Sci ; 23(23): 10564-10574, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31841214

ABSTRACT

OBJECTIVE: Although the natural compound curcumin exerts antitumor properties in vitro, its clinical application is hampered due to rapid metabolism. Light exposure following curcumin application has been demonstrated to improve curcumin's bioavailability. Therefore, this investigation was directed towards evaluating whether light exposure in addition to curcumin application enhances curcumin's efficacy against bladder cancer cell adhesion and migration. MATERIALS AND METHODS: RT112, UMUC3, and TCCSUP cells were incubated with low curcumin concentrations (0.1-0.4 µg/ml) and then exposed to 1.65 J/cm2 visible light for 5 min. Controls remained untreated or were treated with curcumin or light alone. Cell adhesion to Human umbilical vein endothelial cells (HUVECs), to immobilized collagen or fibronectin and chemotactic behavior, integrin α and ß receptor expression with functional relevance, as well as focal adhesion kinase (total and phosphorylated FAK) were evaluated. RESULTS: Curcumin plus light, but neither curcumin nor light alone, significantly altered tumor cell adhesion and suppressed chemotaxis. Integrin α and ß subtypes were dissimilarly modified, depending on the cell line. Suppression of pFAK was noted in RT112 and UMUC3, but not in TCCSUP cells. The integrins α3, α5, and ß1 were involved in curcumin's regulation of adhesion and migration. Blocking studies revealed α3, α5, and ß1 to be associated with TCCSUP adhesion and migration, whereas α5 and ß1, but not α3 contributed to UMUC3 adhesion and migration. Integrin α5 and ß1 controlled RT112 chemotaxis as well, but only α5 was involved in the RT112 adhesion process. CONCLUSIONS: Combining curcumin with light exposure enhances curcumin's anti-tumor potential.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Light , Photochemotherapy/methods , Urinary Bladder Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Biological Availability , Cell Adhesion/drug effects , Cell Adhesion/radiation effects , Cell Culture Techniques , Cell Line, Tumor , Chemotaxis/drug effects , Chemotaxis/radiation effects , Curcumin/therapeutic use , Human Umbilical Vein Endothelial Cells , Humans , Urinary Bladder Neoplasms/pathology
2.
Br J Cancer ; 107(5): 847-55, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22782340

ABSTRACT

BACKGROUND: Inhibitors of the mammalian target of rapamycin (mTOR) might become a novel tool to treat advanced prostate cancer. However, chronic drug exposure may trigger resistance, limiting the utility of mTOR inhibitors. METHODS: Metastatic potential of PC3 prostate cancer cells, susceptible (PC3(par)) or resistant (PC3(res)) to the mTOR-inhibitor RAD001 was investigated. Adhesion to vascular endothelium or immobilised collagen, fibronectin and laminin was quantified. Motility, migration and invasion were explored by modified Boyden chamber assay. Integrin α and ß subtypes were analysed by flow cytometry, western blotting and real-time PCR. Integrin-related signalling, EGFr, Akt, p70S6kinase and ERK1/2 activation were determined. RESULTS: Adhesion was reduced, whereas motility, migration and invasion were enhanced in PC3(res). The α2 and ß1 integrin subtypes were dramatically elevated, integrins α1 and α6 were lowered, whereas α5 was nearly lost in PC3(res). Activation of the Akt signalling pathway was strongly upregulated in these cells. Treating PC3(par) cells with RAD001 reduced motility, migration and invasion and deactivated Akt signalling. Blocking studies revealed that α2 and ß1 integrins significantly trigger the motile behaviour of the tumour cells. CONCLUSION: Chronic RAD001 treatment caused resistance development characterised by distinct modification of the integrin-expression profile, driving prostate cancer cells towards high motility.


Subject(s)
Cell Movement/drug effects , Integrin alpha2/metabolism , Integrin beta1/metabolism , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Everolimus , Humans , Integrin alpha2/biosynthesis , Integrin beta1/biosynthesis , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/biosynthesis
3.
Prostate Cancer Prostatic Dis ; 15(3): 250-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22450844

ABSTRACT

BACKGROUND: The influence of the bisphosphonate zoledronic acid (ZA) on prostate cancer (PC) growth, adhesion and invasive behavior was investigated. METHODS: PC-3, DU-145 and LNCaP cells were treated with ZA, and tumor-cell growth was then investigated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Furthermore, tumor-cell adhesion to vascular endothelium or to immobilized extracellular matrix proteins, as well as migratory properties of the cells, was evaluated. Integrin ß subtypes, integrin-dependent signaling, as well as cell-cycle regulating proteins, were analyzed by western blots. RESULTS: ZA dose-dependently reduced tumor-cell growth but did not impair tumor-endothelium and tumor-matrix interaction. However, ZA significantly inhibited tumor migration and invasive activity. Cyclin E was reduced by ZA in LNCaP and DU-145, and p21 was elevated in LNCaP cells. p27 was upregulated in all tumor cell lines, compared with the controls. ZA elevated ß1-integrin in PC-3 and diminished ß4-integrin in PC-3 and DU-145 cells. CONCLUSIONS: ZA inhibits PC growth and motility but does not influence the mechanical contact between tumor cells and the vascular wall.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Diphosphonates/pharmacology , Imidazoles/pharmacology , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Male , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Zoledronic Acid
4.
World J Urol ; 29(6): 779-86, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20640575

ABSTRACT

PURPOSE: Renal cell carcinoma (RCC) is highly resistant to chemotherapy and unresponsive to radio- and immunotherapy. Recently, we have documented that the histone deacetylase (HDAC)-inhibitor valproic acid (VPA) in combination with low-dosed interferon (IFN)-alpha significantly inhibits RCC proliferation and adhesion in vitro and in vivo. The current study investigated the effects of these compounds on gene transcription of metastatic RCC cell line Caki-1 after 3 and 5 days exposure. METHODS: To evaluate the gene expression profiles of the RCC cells, we performed microarray analysis using Affymetrix GeneChip. Selected significant genes were further validated by Real Time PCR. RESULTS: Microarray revealed that VPA altered genes that are involved in cell growth, cell survival, immune response, cell motility and cell adhesion. Combination of VPA with IFN-alpha not only enhanced the effects on gene transcription but also resulted in the expression of novel genes, which were not induced by either VPA or IFN-alpha alone. Among the up-regulated genes were chemokines (CXCL10, CXCL11, CXCL16) and integrins (ITGA2, ITGA4, ITGA5, ITGA6, ITGA7). Genes encoding for adhesion molecules (NCAM1, ICAM1, VCAM1) were also modulated. Real Time PCR approved these findings. CONCLUSION: This data provides insight into the molecular mechanism of action of the combined treatment of VPA and IFN-alpha in RCC. Implications are that the combined application of VPA and IFN-alpha may represent a more efficient alternative to existing therapy options for RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Interferon-alpha/pharmacology , Kidney Neoplasms/genetics , Valproic Acid/pharmacology , Carcinoma, Renal Cell/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Immunologic Factors/pharmacology , Kidney Neoplasms/pathology , Microarray Analysis
5.
Transplant Proc ; 41(5): 1615-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19545691

ABSTRACT

OBJECTIVE: To evaluate the incidence of voiding dysfunction in older male renal transplant recipients. PATIENTS AND METHODS: Data for 103 patients aged 60 years or older (mean age, 65.7 years; group 1) who underwent transplantation at our center between January 1999 and August 2007 were compared with data for a group of 139 younger patients (mean age, 50.1 years; group 2) treated within the same time frame. RESULTS: Postoperatively, 28 group 1 recipients (27%) and 26 group 2 recipients (19%) experienced voiding dysfunction after removal of the transurethral catheter (P = .12). The most common cause was bladder outlet obstruction due to benign prostatic hyperplasia in 26 patients in group 1 (25%) and 17 patients in group 2 (12%) (P = .009). Bladder neck contracture, urethral stricture, and detrusor underactivity were diagnosed in the other patients. Transurethral resection of the prostate gland was performed in 21 group 1 patients (20%) and 14 group 2 patients (10%) (P = .02) at a mean of 31.1 and 29.5 days, respectively (P = .23) after transplantation. Surgical procedures were performed without complication, and symptoms did not recur postoperatively. CONCLUSIONS: Our data reveal a high incidence of voiding dysfunction in older male renal transplant recipients. High residual urine and urinary retention after renal transplantation may induce recurrent urinary tract infections, cause relevant complications, and seriously affect graft function. Recognizing the substantial effects of postoperative voiding dysfunction will enable optimum management of older kidney transplant recipients.


Subject(s)
Aging/physiology , Kidney Transplantation/adverse effects , Urethral Obstruction/etiology , Urinary Retention/etiology , Urodynamics/physiology , Aged , Humans , Male , Middle Aged , Postoperative Period , Prostatectomy , Prostatic Hyperplasia/epidemiology , Retrospective Studies , Urethral Obstruction/epidemiology , Urethral Obstruction/surgery , Urinary Bladder Neck Obstruction/etiology , Urinary Bladder Neck Obstruction/surgery , Urinary Catheterization , Urinary Retention/epidemiology
6.
Urologe A ; 47(9): 1175-81, 2008 Sep.
Article in German | MEDLINE | ID: mdl-18688594

ABSTRACT

BACKGROUND: Conventional therapeutic approaches to treat advanced renal cell carcinoma (RCC) are of limited benefit. Receptor tyrosine kinase inhibitors (RTKI) may open up novel treatment options. In the present study, the effects of the RTKI AEE788 on the growth and adhesion capacity of RCC cell lines were evaluated in vitro. MATERIALS AND METHODS: RCC cells were treated with AEE788, and alterations of tumor growth and tumor cell interaction with vascular endothelium or extracellular matrix proteins were analyzed. Furthermore, the addition of interferon alpha (IFNalpha) was investigated to see whether it may enhance the anti-tumoral potential of AEE788. RESULTS: AEE788 significantly blocked RCC cell growth and adhesion. Analysis of alpha- and beta-integrins revealed distinct alterations of the receptor expression profile and downregulation of integrin-dependent signaling. Growth-blocking effects were further enhanced when the AEE788-IFNalpha combination protocol was applied. In addition, downregulation of integrin-dependent signaling was more intense in the presence of a combination of AEE788 and IFNalpha than with AEE788 monotherapy. CONCLUSIONS: AEE788 exerts significant anti-tumoral properties, particularly when combined with IFNalpha. AEE788 may therefore be an encouraging compound to treat advanced RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Adhesion/drug effects , Cell Division/drug effects , ErbB Receptors/antagonists & inhibitors , Kidney Neoplasms/pathology , Purines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Humans , In Vitro Techniques , Integrin alpha Chains/metabolism , Integrin beta Chains , Interferon alpha-2 , Interferon-alpha/pharmacology , Recombinant Proteins , Signal Transduction/drug effects
7.
J Cell Mol Med ; 12(6A): 2457-66, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18266964

ABSTRACT

The anti-epileptic drug valproic acid is also under trial as an anti-cancer agent due to its histone deacetylase (HDAC) inhibitory properties. However, the effects of valproic acid (VPA) are limited and concentrations required for exerting anti-neoplastic effects in vitro may not be reached in tumour patients. In this study, we tested in vitro and in vivo effects of two VPA-derivatives (ACS2, ACS33) on pre-clinical prostate cancer models. PC3 and DU-145 prostate tumour cell lines were treated with various concentrations of ACS2 or ACS33 to perform in vitro cell proliferation 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and to evaluate tumour cell adhesion to endothelial cell monolayers. Analysis of acetylated histones H3 and H4 protein expression was performed by western blotting. In vivo tumour growth was conducted in subcutaneous xenograft mouse models. Tumour sections were assessed by immunohistochemistry for histone H3 acetylation and proliferation. ACS2 and ACS33 significantly up-regulated histone H3 and H4 acetylation in prostate cancer cell lines. In micromolar concentrations both compounds exerted growth arrest in PC3 and DU-145 cells and prevented tumour cell attachment to endothelium. In vivo, ACS33 inhibited the growth of PC3 in subcutaneous xenografts. Immunohistochemistry and western blotting confirmed increased histone H3 acetylation and reduced proliferation. ACS2 and ACS33 represent novel VPA derivatives with superior anti-tumoural activities, compared to the mother compound. This investigation lends support to the clinical testing of ACS2 or ACS33 for the treatment of prostate cancer.


Subject(s)
Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors , Prostatic Neoplasms/drug therapy , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Histones/metabolism , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Valproic Acid/analogs & derivatives , Valproic Acid/therapeutic use , Xenograft Model Antitumor Assays
8.
Br J Cancer ; 96(11): 1699-706, 2007 Jun 04.
Article in English | MEDLINE | ID: mdl-17505515

ABSTRACT

Drug resistance to chemotherapy is often associated with increased malignancy in neuroblastoma (NB). In pursuit of alternative treatments for chemoresistant tumour cells, we tested the response of multidrug-resistant SKNSH and of vincristine (VCR)-, doxorubicin (DOX)-, or cisplatin (CDDP)-resistant UKF-NB-2, UKF-NB-3 or UKF-NB-6 NB tumour cell lines to valproic acid (VPA), a differentiation inducer currently in clinical trials. Drug resistance caused elevated NB adhesion (UKF-NB-2(VCR), UKF-NB-2(DOX), UKF-NB-2(CDDP), UKF-NB-3(VCR), UKF-NB-3(CDDP), UKF-NB-6(VCR), UKF-NB-6(CDDP)) to an endothelial cell monolayer, accompanied by downregulation of the adhesion receptor neural cell adhesion molecule (NCAM). Based on the UKF-NB-3 model, N-myc proteins were enhanced in UKF-NB-3(VCR) and UKF-NB-3(CDDP), compared to the drug naïve controls. p73 was diminished, whereas the p73 isoform deltaNp73 was upregulated in UKF-NB-3(VCR) and UKF-NB-3(CDDP). Valproic acid blocked adhesion of UKF-NB-3(VCR) and UKF-NB-3(CDDP), but not of UKF-NB-3(DOX), and induced the upregulation of NCAM surface expression, NCAM protein content and NCAM coding mRNA. Valproic acid diminished N-myc and enhanced p73 protein level, coupled with downregulation of deltaNp73 in UKF-NB-3(VCR) and UKF-NB-3(CDDP). Valproic acid also reverted enhanced adhesion properties of drug-resistant UKF-NB-2, UKF-NB-6 and SKNSH cells, and therefore may provide an alternative approach to the treatment of drug-resistant NB by blocking invasive processes.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Endothelium, Vascular/drug effects , Neuroblastoma/pathology , Valproic Acid/pharmacology , Vincristine/pharmacology , Antigens, Surface/chemistry , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Endothelium, Vascular/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , RNA, Messenger/metabolism , Tumor Cells, Cultured
9.
Int J Clin Pharmacol Ther ; 42(10): 568-74, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15516027

ABSTRACT

The branched-chain fatty acid valproate (valproic acid; VPA) displays antitumoral properties by blocking tumor growth, progression and invasion. Recent data have shown that VPA reduces the angiogenic activity of endothelial cells. The object of this study was to investigate whether endothelial modulation might also influence the level of chemotactic mediators. Endothelial cells were isolated from human umbilical cord veins (HUVEC) and treated with VPA-concentrations ranging from 0.125 mM to 1 mM. The mRNA level of CXC-chemokines was investigated by reverse transcriptase-polymerase chain reaction. The proliferative activity of HUVEC was measured as well. VPA evoked a striking increase in the neutrophil chemoattractants CXCL1, CXCL3, CXCL4, CXCL5 and a moderate increase in CXCL6 with maximal effects after a 3-day incubation period. Other CXC-chemokines and CXC-receptors remained unaffected. HUVEC growth was diminished time- and dose-dependently by VPA. We conclude that VPA treatment leads to alterations in the chemokine expression profile of endothelial cells. This might allow more neutrophils to reach the tumor area and trigger cytolysis.


Subject(s)
Antineoplastic Agents/pharmacology , Chemokines, CXC/biosynthesis , Endothelial Cells/drug effects , Receptors, Chemokine/biosynthesis , Valproic Acid/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Chemokines, CXC/genetics , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Intercellular Signaling Peptides and Proteins , RNA, Messenger/biosynthesis , Receptors, Chemokine/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Clin Exp Immunol ; 134(2): 238-45, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14616783

ABSTRACT

Immunosuppression correlates with the development and recurrence of cancer. Mycophenolate mofetil (MMF) has been shown to reduce adhesion molecule expression and leucocyte recruitment into the donor organ. We have hypothesized that MMF might also prevent receptor-dependent tumour dissemination. Therefore, we have investigated the effects of MMF on tumour cell adhesion to human umbilical vein endothelial cells (HUVEC) and compared them with the effects on T cell-endothelial cell interactions. Influence of MMF on cellular adhesion to HUVEC was analysed using isolated CD4+ and CD8+ T cells, or WiDr colon adenocarcinoma cells as the model tumour. HUVEC receptors ICAM-1, VCAM-1, E-selectin and P-selectin were detected by flow cytometry, Western blot or Northern blot analysis. Binding activity of T cells or WiDr cells in the presence of MMF were measured using immobilized receptor globulin chimeras. MMF potently blocked both T cell and WiDr cell binding to endothelium by 80%. Surface expression of the endothelial cell receptors was reduced by MMF in a dose-dependent manner. E-selectin mRNA was concurrently reduced with a maximum effect at 1 microm. Interestingly, MMF acted differently on T cells and WiDr cells. Maximum efficacy of MMF was reached at 10 and 1 microm, respectively. Furthermore, MMF specifically suppressed T cell attachment to ICAM-1, VCAM-1 and P-selectin. In contrast, MMF prevented WiDr cell attachment to E-selectin. In conclusion, our data reveal distinct effects of MMF on both T cell adhesion and tumour cell adhesion to endothelial cells. This suggests that MMF not only interferes with the invasion of alloactivated T cells, but might also be of value in managing post-transplantation malignancy.


Subject(s)
Colonic Neoplasms/pathology , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion/drug effects , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Colonic Neoplasms/immunology , Dose-Response Relationship, Immunologic , Down-Regulation/drug effects , Endothelium, Vascular/immunology , Humans , Integrins/drug effects , Integrins/metabolism , Tumor Cells, Cultured
11.
Spinal Cord ; 41(11): 610-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14569262

ABSTRACT

STUDY DESIGN: In vitro study on the effects of mycophenolate mofetil (MMF) on isolated human monocytes and endothelial cells. OBJECTIVES: Haematogenous macrophages play an essential role in the development of secondary damage following spinal cord injury (SCI), and there is evidence that the use of immunosuppressants such as MMF can reduce monocyte invasion and neuronal damage. SETTING: University Hospital for Orthopaedic Surgery, Frankfurt am Main, Germany. METHODS: The effects of MMF on the adhesion of human monocytes to human umbilical vein endothelial cells (HUVEC), monocyte binding to immobilised E-selectin, and monocyte expression of intercellular adhesion molecule (ICAM)-1, sialyl Lewis X (sLeX) and major histocompatibility complex (MHC)-II were studied. The binding of monocytes to E-selectin was examined by using purified and immobilised E-selectin fusion protein. Adhesion molecule expression was investigated by flow cytometry. RESULTS: The binding of monocytes to HUVEC was significantly reduced by 30.1% after treatment of monocytes with MMF (10 microg/ml), whereas the pretreatment of HUVEC with MMF did not result in significant changes in monocyte adhesion. MMF forcefully inhibited monocyte binding to immobilised E-selectin by 55.7%. Furthermore, MMF significantly inhibited the upregulation of ICAM-1- and MHC-II-expression on monocytes stimulated with either lipopolysaccharide or interferon-gamma, whereas the expression of sLeX was not impaired. Toxic effects were excluded by propidium-iodide staining and measurement of fluorescein-diacetate metabolism. CONCLUSION: MMF can downregulate important monocytic adhesion molecules and inhibits monocyte adhesion to endothelial cells, thus indicating that treatment with MMF could be beneficial after SCI. SPONSORSHIP: This study was supported by the DFG (Ha 2721/1-3), the Paul und Ursula Klein-Stiftung and the Stiftung Friedrichsheim.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Communication/drug effects , Endothelial Cells/drug effects , Immunosuppressive Agents/pharmacology , Monocytes/drug effects , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Survival , Cells, Cultured , Chromatography, High Pressure Liquid , Cytotoxicity Tests, Immunologic/methods , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Interactions , E-Selectin/metabolism , Endothelial Cells/metabolism , Flow Cytometry/instrumentation , Flow Cytometry/methods , Humans , Immunohistochemistry/methods , In Vitro Techniques , Intercellular Adhesion Molecule-1/metabolism , Lewis X Antigen/metabolism , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Propidium/metabolism , Transcription Factors , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism
12.
Zentralbl Chir ; 128(4): 278-82, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12700983

ABSTRACT

OBJECTIVE: To evaluate the impact of passenger leukocytes in liver grafts on the rate of microchimerism induction after liver transplantation and to evaluate immunological changes thereafter based on serial donor-specific MLC's in these patients. METHODS: 26 orthotopic liver transplant recipients were prospectively evaluated for immunological changes based on the co-transplantation of donor-derived leukocytes. Intraoperatively harvested liver biopsies and peripheral blood-lymphocytes of liver transplant recipients were sampled at various time points. Donor spleen cells were obtained during organ procurement. RESULTS: HLA-PCR analysis demonstrated a stable pattern of microchimerism in 15 out of 26 patients. Microchimerism was detectable by PCR up to a mean of 7 weeks after transplantation, when chimerism in the peripheral blood became negative. Passenger donor leukocytes were present in all biopsies obtained during backtable preparation of the liver graft. For the 15 patients presenting microchimerism the rate of passenger leukocytes in the liver graft biopsies showed a mean of 155.8 leukocytes per mm 2 liver tissue (SD +/- 23.2 cells/mm2, range 121 to 217 cells per mm2 tissue). Otherwise patients without chimerism showed a mean of 90.4 passenger leukocytes per mm2 tissue (SD +/- 14.5 cells/mm2, range: 52 to 99 cells/mm2). Lymphocyte proliferation, determined by donor-specific "multiple" single-way- mixed-lymphocyte-cultures (dsmMLC) was reduced to a mean of 62.2 % of preoperative values (SD +/- 14.5 %, range 33 % to 88 %) in the 15 patients with stable microchimerism. Otherwise in the 11 patients without microchimerism dsmMLC results stayed at continuously higher levels with a mean of 106 % (SD +/- 13.4, range 92 % to 134 %). CONCLUSIONS: The results from these studies of microchimerism and lymphocyte reactivity after liver transplantation suggest that the co-transplantation of donor leukocytes plays an important and active role in the modulation of the host-immune system.


Subject(s)
Leukocytes , Liver Transplantation/methods , Lymphocyte Culture Test, Mixed , Transplantation Chimera/immunology , Biopsy, Needle , Follow-Up Studies , HLA-DR Antigens/genetics , Humans , Leukocytes/immunology , Leukocytes/pathology , Liver/pathology , Liver Transplantation/immunology , Liver Transplantation/pathology , Lymphocyte Activation/immunology , Phenotype , Polymerase Chain Reaction , Polymorphism, Genetic , Tissue Donors , Transplantation Tolerance/immunology
13.
Zentralbl Chir ; 128(4): 283-90, 2003 Apr.
Article in German | MEDLINE | ID: mdl-12700984

ABSTRACT

Over the past few years, hepatocyte transplantation has been considered as an alternative method for orthotopic liver transplantation for the treatment of various liver diseases. Beside curative approach for genetic metabolic deficiencies (familial hypercholesterolemia, hemophilia, etc.), it could be a useful tool for bridging the waiting period until an appropriate donor organ is obtained. In preclinical animal studies, hepatocytes injected intraperitoneally, intraportally or into the spleen settle down in the diseased liver. This enables genetic modification to correct inborn metabolic deficiencies and improves survival in acute liver failure. In 1992, the first clinical transplantation of isolated hepatocytes in 10 patients was performed. In 1998, Fox and coworkers described the successful transplantation of allogeneic liver cells in a child with Crigler-Najjar syndrome. Accomplished studies of Strom et al. resp. Bilir et al. of the same year proved the effectiveness of liver cell transplantation for transient treatment of acute liver failure. Prerequisite of this cell-based therapeutic strategy is a sufficient amount of highly differentiated hepatocytes, hence, a well established in-vitro cell-culture technique is necessary to yield a reproducible number of proliferating hepatocytes and to preserve the physiological cell function. This review discusses the different experimental approaches regarding the cultivation of human hepatocytes and also the use of alternative cell sources (like animal hepatocytes, immortalized cells of human origin, progenitor cells from fetal human liver/liver stem cells) for hepatocyte transplantation.


Subject(s)
Hepatocytes/transplantation , Liver Failure/surgery , Metabolism, Inborn Errors/surgery , Animals , Cells, Cultured , Humans , Liver Failure/genetics , Metabolism, Inborn Errors/genetics , Treatment Outcome
14.
Antiviral Res ; 55(1): 179-88, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12076762

ABSTRACT

Antiviral activity of the metal chelator ethylenediaminedisuccinic acid (EDDS) was examined in vitro against human cytomegalovirus (HCMV) wild type strains and strains that are resistant against ganciclovir (GCV) and cidofovir (HPMPC). EDDS inhibited the replication of wild-type as well as GCV- and HPMPC-resistant strains with a 50% effective concentration of 7.4-12 microg/ml. At concentrations of 100 microg/ml EDDS, unlike GCV or HPMPC, suppressed HCMV-induced up-regulation of intercellular adhesion molecule-1 (ICAM-1) and reduced T-cell adhesion to HCMV-infected cells in a monolayer adhesion model. In vitro EDDS inhibited murine cytomegalovirus (MCMV) replication (EC50 8.6 microg/ml) and caused in mice some protection against MCMV induced mortality at a non-toxic dose. Since immunopathological factors may play a significant role in HCMV disease it will be of interest to further study whether EDDS is effective in terms of modulation of inflammatory responses to HCMV infections.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antiviral Agents/pharmacology , Chelating Agents/pharmacology , Cytomegalovirus/drug effects , Cytosine/analogs & derivatives , Ethylenediamines/pharmacology , Organophosphonates , Succinates/pharmacology , Animals , Cell Adhesion/drug effects , Cell Line , Cidofovir , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Cytosine/pharmacology , Drug Resistance, Microbial , Herpesviridae Infections/drug therapy , Herpesviridae Infections/mortality , Humans , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , Mice , Mice, SCID , Muromegalovirus/drug effects , Organophosphorus Compounds/pharmacology , Virus Replication/drug effects
15.
Hepatology ; 33(3): 519-29, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11230730

ABSTRACT

Although the control of biliary ductular morphogenesis has received some attention particularly using isolated rat biliary epithelial cell models, the regulation of human bile duct formation is not well defined. In the present study, using a 3-dimensional culture model comprising primary human biliary epithelial cells (BECs) and coculture with primary human hepatocytes, we have sought to define the factors involved. We have shown that primary human BECs can be expanded on collagen gels in the absence of growth factors or serum. When plated in high density in double collagen gels, BECs established 3-dimensional structures that subsequently developed into well differentiated polarized luminal ducts. This morphogenic response occurred in the absence of hepatocyte growth factor (HGF) and epidermal growth factor. Strikingly, the addition of growth factors (in the presence of serum) resulted in loss of polarity although the cells retained growth responses to both factors. Coculture of BECs with autologous human hepatocytes enhanced the ability of low-density BECs to undergo ductulogenesis. This effect was mimicked by addition of conditioned medium from previous hepatocyte-BEC cocultures. These findings indicate that for human biliary ductular morphogenesis, epithelial cell-cell interactions are required but that mesenchymally derived factors such as HGF may not be important.


Subject(s)
Bile Ducts/cytology , Cytological Techniques , Hepatocytes/physiology , Bile Ducts/physiology , Cell Division/drug effects , Cell Polarity/drug effects , Cells, Cultured , Coculture Techniques , Collagen , Culture Media, Conditioned , Culture Media, Serum-Free , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/physiology , Gels , Hepatocyte Growth Factor/pharmacology , Humans
16.
Neurosci Lett ; 298(1): 33-6, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11154829

ABSTRACT

Several factors contribute to the maintenance of central nervous system immune privilege and astrocytes have been identified as a major source of immunomodulatory cytokines. To investigate whether hematogenous monocytes are immunologically deactivated by astrocyte-derived factors human monocytes were stimulated with lipopolysaccharide or interferon (IFN)-gamma and treated with the supernatant from pure astrocyte cultures, interleukin (IL)-4, IL-10, or with IL-1-receptor antagonist (1L-1-RA). Flow cytometry demonstrated that the supernatant from astrocyte cultures was the most potent agent in reducing the levels of major histocompatibility complex (MHC)-class-II- as well as intercellular adhesion molecule-1-expression, whereas IL-4, IL-10, and IL-1-RA had only marginal effects. The expression of leukocyte function antigen-1 and very late antigen-4 was not modulated by either factor. In conclusion, astrocytes seem to provide soluble factors that have the capacity to deactivate hematogenous monocytes.


Subject(s)
Astrocytes/metabolism , Histocompatibility Antigens Class II/metabolism , Intercellular Adhesion Molecule-1/metabolism , Monocytes/metabolism , Cells, Cultured , Down-Regulation , Flow Cytometry , Humans , Integrin alpha4beta1 , Integrins/metabolism , Interferon-gamma/pharmacology , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/pharmacology , Interleukin-10/metabolism , Interleukin-4/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Function-Associated Antigen-1/metabolism , Receptors, Lymphocyte Homing/metabolism , Sialoglycoproteins/metabolism
17.
Transplantation ; 70(1): 236-40, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919613

ABSTRACT

Interaction of endothelial P-selectin with sialyl Lewis(x)-glycoprotein or P-selectin glycoprotein ligand (PSGL)-1 on leukocytes represents an early step in leukocyte recruitment. Redistribution of P-selectin to the endothelial cell surface occurs rapidly after challenge with several proinflammatory agents, for example, histamine, leucopterins, or lipopolysaccharide. We present evidence that prostaglandin E2 (PGE2) is an efficient inductor of surface P-selectin on cultured human umbilical vein endothelial cells (HUVEC). The increase in P-selectin-immunoreactivity coincided with redistribution of cytoplasmic P-selectin-reactive granulae to the endothelial cell surface, as visualized by confocal laser microscopic examination. CD4-T-cell adhesion to PGE2-stimulated HUVEC was also enhanced by a factor of 4, and blocking mAb directed against the binding site of P-selectin almost completely abrogated this increase in CD4-T-cell adhesion. In summary, our findings show that liberation of PGE2 is an important inductor of P-selectin surface expression on endothelial cells, resulting in enhanced recruitment of inflammatory cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Dinoprostone/pharmacology , Endothelium, Vascular/cytology , P-Selectin/biosynthesis , Cell Adhesion , Cells, Cultured , Endothelium, Vascular/metabolism , Histamine/pharmacology , Humans , Tetradecanoylphorbol Acetate/pharmacology , Umbilical Veins/cytology
18.
Tissue Antigens ; 55(5): 412-21, 2000 May.
Article in English | MEDLINE | ID: mdl-10885561

ABSTRACT

Previously, experimental in vivo results showed that the productively and persistently human cytomegalovirus (HCMV)-infected neuroblastoma cell line UKF-NB-4AD169 exhibits a more malignant phenotype than the non-infected variant UKF-NB-4. To prove the assumption that enhanced malignancy may be due to enhanced invasive potential of the infected cells we studied interactions of both lines with monolayers of cultured endothelial cells. UKF-NB-4AD169 cells adhered to and transmigrated through endothelial monolayer to a significantly higher extent compared with UKF-NB4. Furthermore, the adhesion of UKF-NB-4AD169 but not of UKF-NB4 resulted in focal disruption of the monolayer integrity which facilitates tumor cell transmigration. Blocking antibodies directed against the beta1 integrin chain as well as beta1alpha5 on the tumor cells specifically inhibited adhesion in a concentration-dependent manner. When UKF-NB-4 were pretreated with a beta1 integrin activating antibody, focal disruption of the endothelial integrity also occurred. These findings lead us to suggest that HCMV infection activates beta1alpha5 in the host neuroblastoma cell which in turn enables these cells to tightly adhere to endothelial cells. In the presence of the protease inhibitor phenantroline, beta1alpha5-mediated adhesion was not impaired whereas UKF-NB4AD169-mediated endothelial monolayer permeabilization was dose dependently inhibited. We conclude that human cytomegalovirus infection contributes to augmented neuroblastoma invasiveness via adhesion of activated beta1alpha5 and subsequent matrix digestion by proteases.


Subject(s)
Cytomegalovirus Infections/pathology , Cytomegalovirus , Neuroblastoma/pathology , Receptors, Fibronectin/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Adhesion/physiology , Cell Communication/physiology , Cell Movement/physiology , E-Selectin/analysis , Epithelial Cells/chemistry , Epithelial Cells/cytology , Epithelial Cells/virology , Flow Cytometry , Humans , Immunophenotyping , Intercellular Adhesion Molecule-1/analysis , Membrane Glycoproteins/analysis , Neoplasm Invasiveness , Neuroblastoma/virology , Receptors, Fibronectin/analysis , Receptors, Fibronectin/immunology , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/virology , Umbilical Cord/cytology , Vascular Cell Adhesion Molecule-1/analysis
19.
Transplantation ; 69(9): 1977-81, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10830246

ABSTRACT

BACKGROUND: Prostaglandin E2 (PGE2) is a powerful endogenous immune suppressant and interferes with various T-cell functions. However, it is not known in detail whether immunosuppressive drugs influence the PGE2-driven immune response in transplant patients. Therefore, we investigated the effect of several immunosuppressive compounds, in particular the novel drug mycophenolate mofetil (MMF), on endothelial PGE2 release. METHODS: Endothelial cells (HUVEC) were activated by either allogeneic CD4+ or CD8+ T cells, or by the cytokines interleukin-1 or gamma-interferon. Using an enzyme-linked immunosorbent assay, we analyzed PGE2 release of the activated HWEC in the presence of MMF, cyclosporine, or tacrolimus. As verapamil and mibefradil also possess immunosuppressive properties, they were included in the study as well. RESULTS: Activation of HUVEC with interleukin-1 or T cells resulted in a drastic accumulation of PGE2 in the supernatant. Cyclosporine or tacrolimus had no effect on PGE2 release. However, Ca2+ channel blockers, when applied at higher dosages, caused a significant increase in PGE2. Interestingly, MMF strongly diminished the PGE2 level in the cell culture supernatant in a concentration-dependent manner. CONCLUSION: The results demonstrate an inhibitory effect of MMF on PGE2 production, which may lower the benefits of the PGE2-triggered immune response after organ transplantation.


Subject(s)
Cytokines/pharmacology , Dinoprostone/biosynthesis , Endothelium, Vascular/metabolism , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/analogs & derivatives , T-Lymphocytes/physiology , Cells, Cultured , Cyclosporine/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Mycophenolic Acid/pharmacology , Tacrolimus/pharmacology
20.
Transplantation ; 69(4): 588-97, 2000 Feb 27.
Article in English | MEDLINE | ID: mdl-10708116

ABSTRACT

BACKGROUND: Cyclosporine A (CsA) and tacrolimus prevent proliferation but not transendothelial migration of alloreactive lymphocytes into donor organs. As a result, serious adverse effects, such as nephrotoxicity and neurotoxicity, have been observed under CsA/tacrolimus therapy. The incorporation of new drugs with infiltration blocking properties might enhance the efficacy of the current immunosuppressive protocol, allowing lower CsA/tacrolimus dosage. Because Ca2+ plays a critical role in cell-cell interaction, the Ca2+-channel blocker verapamil might be a good cany. didate for supporting CsA/tacrolimus-based therapy. METHODS: A T-cell endothelial cell coculture model or immobilized immunoglobulin G globulin chimeras were employed to investigate how S- and R- verapamil interfere with the lymphocytic infiltration process. The expression and arrangement of membranous adhesion receptors and cytoskeletal F-actin filaments were analyzed by fluorometric method in the presence of. verapamil. RESULTS: Both verapamil enantiomers strongly inhibited lymphocyte infiltration. CD4+ and CD8+ T-cells were influenced to a similar extent with regard to horizontal locomotion (CD4+=CD8+), but to a different extent with regard to adhesion and penetration (CD4+ > CD8+). Moreover, penetration was blocked to a higher extent than was adhesion. ID50-values were 31 microM (CD4+-adhesion) and 11 microM (CD4+-penetration). Verapamil reduced P-selectin expression on endothelial cells and effectively down-regulated binding of T-cells to immobilized P-selectin immunoglobulin G globulins (ID50=4.4 microM; CD4+). A verapamil-induced reduction of intracellular F-actin in T-lymphocytes was proven to be mainly responsible for diminished cell locomotion. CONCLUSIONS: The prevention of CD4+ T-cell penetration by verapamil might argue for its use as an adjunct to CsA/tacrolimus-based immunosuppressive therapy.


Subject(s)
Immune Tolerance/drug effects , Verapamil/pharmacology , Cell Movement/drug effects , Dinoprostone/pharmacology , E-Selectin/biosynthesis , E-Selectin/drug effects , Endothelium, Vascular/cytology , Humans , Intercellular Adhesion Molecule-1/drug effects , Lewis X Antigen/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , P-Selectin/biosynthesis , P-Selectin/drug effects , Protein Binding , T-Lymphocytes/cytology , Vascular Cell Adhesion Molecule-1/drug effects , Verapamil/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...