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










Publication year range
1.
Int J Mol Sci ; 20(9)2019 May 13.
Article in English | MEDLINE | ID: mdl-31086078

ABSTRACT

Previously, we have reported that the active vitamin D metabolite, calcitriol and vitamin D3 (cholecalciferol), both remarkably inhibit hepatitis C virus production. The mechanism by which vitamin D3 exerts its effect is puzzling due to the low levels of calcitriol produced in vitamin D3-treated Huh7.5 cells. In this study, we aimed to explore the mechanism of vitamin D3 anti-hepatitis C virus effect. We show that vitamin D3 activity is not mediated by its metabolic conversion to calcitriol, but may be due to its primary metabolic product 25(OH)D3. This is inferred from the findings that 25(OH)D3 could inhibit hepatitis C virus production in our system, and that adequate concentrations needed to exert this effect are produced in Huh7.5 cells treated with vitamin D3. Using the CRISPR-Cas9 editing technology to knockout the vitamin D receptor, we found that the antiviral activity of vitamin D3 and 25(OH)D3 was not impaired in the vitamin D receptor knockout cells. This result indicates that 25(OH)D3 anti-hepatitis C virus effect is exerted by a vitamin D receptor-independent mode of action. The possibility that vitamin D3 and 25(OH)D3, being 3ß-hydroxysteroids, affect hepatitis C virus production by direct inhibition of the Hedgehog pathway in a vitamin D receptor-independent manner was ruled out. Taken together, this study proposes a novel mode of action for the anti-hepatitis C virus activity of vitamin D3 that is mediated by 25(OH)D3 in a vitamin D receptor-independent mechanism.


Subject(s)
Calcifediol/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Hepacivirus/drug effects , Hepacivirus/physiology , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Receptors, Calcitriol/metabolism , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cholecalciferol/pharmacology , Humans , Real-Time Polymerase Chain Reaction , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
NPJ Breast Cancer ; 3: 10, 2017.
Article in English | MEDLINE | ID: mdl-28649650

ABSTRACT

Radiation dermatitis occurs frequently during adjuvant radiation therapy for breast cancer. Prevention of radiation dermatitis by applying various creams and ointments has a limited success, and Aqua cream which has urea as one of its active ingredients is used in many institutions as a preventive treatment. The primary goal of this study is to assess the effect of vitamin D (calcipotriol) ointment in prevention of radiodermatitis in breast cancer patients compared to Aqua cream. Twenty-three women with localized breast cancer who underwent breast-conserving surgery and opted to receive adjuvant radiotherapy to breast only were enrolled in this study. A cream containing an active vitamin D analog, calcipotriol (Daivonex), was randomly applied either to the medial or to the lateral half of the irradiated breast, while Aqua cream was applied to the complimentary half of the same breast along the whole treatment days, each day, after the delivery of radiation. Skin reaction was recorded and compared between the two halves of the breast. Vitamin D was well tolerated by patients with no local or systemic allergic reactions. Radiation dermatitis was not significantly different between both treatment arms. Topical vitamin D ointment is not superior to Aqua cream for prevention of radiation-induced dermatitis in women treated with adjuvant radiation for breast cancer.

3.
Dermatoendocrinol ; 8(1): e1137399, 2016.
Article in English | MEDLINE | ID: mdl-27195054

ABSTRACT

Several inflammatory mediators increase calcitriol production by epidermal keratinocytes. In turn calcitriol attenuates the keratinocyte inflammatory response. Since the effect of the in-situ generated calcitriol depends also on the sensitivity to the hormone we studied the effect of inflammatory cytokines on the response of HaCaT human keratinocytes to calcitriol by examining the expression and transcriptional activity of VDR. Treatment with TNF, but not with IL-1ß or interferon γ, increased VDR protein level, while decreasing the level of its heterodimerization partner RXRα. This was associated with increased VDR mRNA levels. c-Jun N-terminal kinase, but not P38 MAPK or NFκB, was found to participate in the upregulation of VDR by TNF. The functional significance of the modulation of VDR and RXRα levels by TNF is manifested by increased induction of VDR target gene CYP24A1 by calcitriol. Calcitriol, in turn, inhibited the enhanced expression of VDR by TNF. In conclusion, the inflammatory cytokine TNF increases the response of keratinocytes to calcitriol through upregulation of its receptor VDR, which in turn is subject to negative feedback by the hormone accelerating the return of the keratinocyte vitamin D system to its basal activity. We surmise that the increased generation and sensitivity to calcitriol in keratinocytes play a role in the resolution of epidermal inflammation.

4.
J Cell Physiol ; 231(4): 837-43, 2016 04.
Article in English | MEDLINE | ID: mdl-26280673

ABSTRACT

The active metabolite of vitamin D calcitriol and its analogs are well-known for their anti-inflammatory action in the skin, while their main side effect associated with topical treatment of inflammatory disorders is irritant contact dermatitis. Prostaglandin E2 (PGE2 ) is pro-inflammatory at the onset of inflammation and anti-inflammatory at its resolution. We hypothesized that induction of PGE2 synthesis by calcitriol in epidermal keratinocytes may contribute both to its pro-inflammatory and anti-inflammatory effects on the skin. Treatment of human immortalized HaCaT keratinocytes with calcitriol (3-100 nM, 2-24 h) increased PGE2 production due to increased mRNA and protein expression of COX-2, but not to increase of COX-1 or release of arachidonic acid. The effect of calcitriol on COX-2 mRNA was observed also in primary human keratinocytes. The increase in COX-2 mRNA is associated with COX-2 transcript stabilization. Calcitriol exerts this effect by a rapid (2 h) and protein synthesis independent mode of action that is dependent on PKC and Src kinase activities. Treatment with a COX-2 inhibitor partially prevented the attenuation of the keratinocyte inflammatory response by calcitriol. We conclude that upregulation of COX-2 expression with the consequent increase in PGE2 synthesis may be one of the mechanisms explaining the Janus face of calcitriol as both a promoter and attenuator of cutaneous inflammation. J. Cell. Physiol. 231: 837-843, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Keratinocytes/metabolism , Vitamin D/pharmacology , Anti-Inflammatory Agents/pharmacology , Arachidonic Acid/metabolism , Calcitriol/pharmacology , Cell Line , Cells, Cultured , Cyclooxygenase 2/genetics , Humans , Protein Kinase C/metabolism , RNA Stability/drug effects , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , src-Family Kinases/metabolism
5.
J Cell Physiol ; 227(5): 2175-83, 2012 May.
Article in English | MEDLINE | ID: mdl-21792935

ABSTRACT

The hormonal form of vitamin D, calcitriol, and its analogs are known for their beneficial effect in the treatment of inflammatory skin disorders. Keratinocytes play a role in epidermal inflammatory responses invoked by breeching of the epidermal barrier, by infectious agents and by infiltrating immune cells. We studied the role of calcitriol in the initiation of keratinocyte inflammatory response by the viral and injury mimic polyinosinic-polycytidylic acid (poly(I:C)) and in its maintenance by tumor-necrosis-factor α (TNFα) and investigated the role of the mitogen-activated protein kinase cascades in these processes and their regulation by calcitriol. The inflammatory response of human HaCaT keratinocytes to poly(I:C) or TNFα was assessed by measuring mRNA levels of 13 inflammation-related molecules by real-time PCR microarray and by in-depth investigation of the regulation of interleukin 8, intercellular-adhesion-molecule 1, and TNFα expression. We found that while calcitriol had only a minor effect on the keratinocyte response to poly(I:C) and a modest effect on the early response (2 h) to TNFα, it markedly attenuated the later response (16-24 h) to TNFα. The expression of CYP27B1, the enzyme responsible for calcitriol production, was marginally increased by poly(I:C) and markedly by TNFα treatment. This pattern suggests that while allowing the initial keratinocyte inflammatory response to proceed, calcitriol contributes to its timely resolution. Using pharmacological inhibitors we found that while the p38 MAPK and the extracellular signal-regulated kinase have only a minor role, c-Jun N-terminal kinase plays a pivotal role in the induction of the pro-inflammatory genes and its modulation by calcitriol.


Subject(s)
Keratinocytes/drug effects , Keratinocytes/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Vitamin D/pharmacology , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Cell Line , Enzyme Inhibitors/metabolism , Gene Expression/immunology , Gene Expression Profiling , Humans , Intercellular Adhesion Molecule-1/immunology , Interferon Inducers/immunology , Interleukin-8/immunology , Keratinocytes/cytology , Microarray Analysis , Mitogen-Activated Protein Kinases/metabolism , Poly I-C/immunology , Tumor Necrosis Factor-alpha/immunology , Vitamins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Hepatology ; 54(5): 1570-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21793032

ABSTRACT

UNLABELLED: Vitamin D supplementation was reported to improve the probability of achieving a sustained virological response when combined with antiviral treatment against hepatitis C virus (HCV). Our aim was to determine the in vitro potential of vitamin D to inhibit HCV infectious virus production and explore the mechanism(s) of inhibition. Here we show that vitamin D(3) remarkably inhibits HCV production in Huh7.5 hepatoma cells. These cells express CYP27B1, the gene encoding for the enzyme responsible for the synthesis of the vitamin D hormonally active metabolite, calcitriol. Treatment with vitamin D(3) resulted in calcitriol production and induction of calcitriol target gene CYP24A1, indicating that these cells contain the full machinery for vitamin D metabolism and activity. Notably, treatment with calcitriol resulted in HCV inhibition. Collectively, these findings suggest that vitamin D(3) has an antiviral activity which is mediated by its active metabolite. This antiviral activity involves the induction of the interferon signaling pathway, resulting in expression of interferon-ß and the interferon-stimulated gene, MxA. Intriguingly, HCV infection increased calcitriol production by inhibiting CYP24A1 induction, the enzyme responsible for the first step in calcitriol catabolism. Importantly, the combination of vitamin D(3) or calcitriol and interferon-α synergistically inhibited viral production. CONCLUSION: This study demonstrates for the first time a direct antiviral effect of vitamin D in an in vitro infectious virus production system. It proposes an interplay between the hepatic vitamin D endocrine system and HCV, suggesting that vitamin D has a role as a natural antiviral mediator. Importantly, our study implies that vitamin D might have an interferon-sparing effect, thus improving antiviral treatment of HCV-infected patients.


Subject(s)
Calcitriol/biosynthesis , Cholecalciferol/pharmacokinetics , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Hepatocytes , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Calcitriol/metabolism , Carcinoma, Hepatocellular , Cell Line, Tumor , Drug Synergism , Hepacivirus/growth & development , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Interferon-alpha/pharmacology , Liver Neoplasms , Signal Transduction/drug effects , Signal Transduction/physiology , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Vitamin D3 24-Hydroxylase , Vitamins/pharmacokinetics
7.
Transplantation ; 92(1): 18-24, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21516061

ABSTRACT

BACKGROUND: Liver transplantation is the only therapy of proven benefit in fulminant hepatic failure (FHF). Lipopolysaccharide (LPS), d-galactosamine (GalN)-induced FHF is a well-established model of liver injury in mice. Erythropoietin has a powerful tissue-protective effect in animal models. The aim of this study was to investigate the effect and mechanism of recombinant human erythropoietin (rhEPO) administration in FHF mice. METHODS: C57BL/6 (n=42) mice were studied in vivo in a fulminant model induced by GalN/LPS. rhEPO was administered 30 min after the induction of FHF. Serum liver enzymes and hepatic tumor necrosis factor (TNF)-α and interleukin (IL)-1ß levels were determined. Histologic analysis was performed, and apoptotic cells were identified by immunohistochemistry for caspase-3. Nuclear factor (NF)-κB and c-Jun-N-terminal kinase (JNK) activation were studied using Western blot analysis. RESULTS: After the induction of FHF, all control mice died within 12 hr of GalN/LPS administration. However, 83% of mice that were administered rhEPO were alive 2 weeks later, and overall survival improved (Kaplan-Meier, P<0.001). The serum liver enzymes, hepatic TNF-α and IL-1ß levels, liver histologic injury, and apoptotic hepatocytes were significantly reduced in FHF mice that were administered rhEPO compared with untreated mice. A significant decrease in hepatic NF-κB and JNK activation was noted in FHF rhEPO-treated mice compared with FHF untreated mice. CONCLUSIONS: The administration of rhEPO brought about increased survival and attenuation of the hepatic injury. This was associated with decreased hepatic NF-κB and JNK activation and thus TNF-α and IL-1ß levels. These findings have important implications for the potential use of rhEPO in FHF.


Subject(s)
Erythropoietin/pharmacology , Liver Failure, Acute/drug therapy , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Disease Models, Animal , Galactosamine/toxicity , Humans , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Kaplan-Meier Estimate , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Recombinant Proteins , Tumor Necrosis Factor-alpha/metabolism
8.
Can J Physiol Pharmacol ; 88(12): 1130-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21164559

ABSTRACT

Ischemia-reperfusion injury (I/R) is the main cause of primary graft nonfunction. Our aim was to evaluate the effect of excessive versus acute administration of erythropoietin (EPO) in attenuating the hepatic injury induced by I/R in mice. The effect of segmental (70%) hepatic ischemia was evaluated in a transgenic mouse line with constitutive overexpression of human EPO cDNA and in wild-type (WT) mice. Mice were randomly allocated to 5 main experimental groups: (i) WT-sham, (ii) WT ischemia, (iii) WT ischemia + recombinant human erythropoietin (rhEPO), (iv) transgenic-sham, and (v) transgenic ischemia. The EPO-pretreated mice showed a significant reduction in liver enzyme levels and intrahepatic caspase-3 activity and fewer apoptotic hepatocytes (p < 0.05 for all) compared with the WT untreated I/R group. EPO decreased c-Jun N-terminal kinase (JNK) phosphorylation and nuclear factor-κB (NF-κB) expression during I/R. In transgenic I/R livers, baseline histology showed diffused hepatic injury, and no significant beneficial effect was noted between the WT untreated and the transgenic I/R mice. In conclusion, acute pretreatment with EPO in WT mice attenuated in vivo I/R liver injury. However, in excessive EPO overexpression, the initial liver injury abolished the beneficial effect of EPO. These findings have important implications for the potential use of acute EPO in I/R injury during liver transplantation.


Subject(s)
Erythropoietin/administration & dosage , Liver/blood supply , Reperfusion Injury/drug therapy , Animals , Apoptosis/drug effects , Caspase 3/metabolism , DNA, Complementary/genetics , Female , Hematocrit , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Phosphorylation , Random Allocation , Recombinant Proteins , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
9.
Liver Transpl ; 16(5): 631-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20440772

ABSTRACT

The only currently offered curative option for many patients with primary or secondary liver tumors is the resection of hepatic tumors. The aim of this study was to evaluate the role of recombinant human erythropoietin (rhEPO) in liver protection and regeneration after subtotal hepatectomy in rats. Rats undergoing 70% hepatectomy received an intraperitoneal injection of saline (control) or rhEPO (4 U/g) 30 minutes prior to resection. Liver function was assessed by the measurement of the international normalized ratio (INR) levels, and hepatic injury was assessed by serum alanine aminotransferase and aspartate aminotransferase levels. Hepatic apoptosis was assessed by intrahepatic caspase-3 activity and morphological criteria. The regeneration capacity of remnant livers was assessed over 7 days with the regenerated liver/body weight ratio, immunohistochemistry markers of cell proliferation (Ki-67) and angiogenesis (von Willebrand factor), and phosphorylated extracellular signal-regulated kinase signaling. Two and 4 days after subtotal hepatectomy, the regenerated liver/body weight ratio was significantly higher in animals treated with rhEPO versus the control group (P < 0.005). Serum liver enzymes and INR levels on days 2 and 4 post-hepatectomy were significantly lower in animals pretreated with rhEPO in comparison with the control group (P < 0.005). No statistically significant difference was noted in intrahepatic hepatic caspase-3 activity, immunohistochemistry for caspase-3, or a terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assay between the hepatectomized groups. In the rhEPO-pretreated group, the mitotic index, Ki-67 and von Willebrand factor expression, and extracellular signal-regulated kinase activity were significantly higher on day 2 post-hepatectomy (P < 0.05) in comparison with the control group. In conclusion, rhEPO treatment may offer a unique beneficial dual-function strategy for hepatic protection and regeneration immediately after subtotal hepatectomy in rats.


Subject(s)
Erythropoietin/pharmacology , Hepatectomy/methods , Liver Regeneration/drug effects , Postoperative Complications/drug therapy , Reperfusion Injury/drug therapy , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Blotting, Western , Caspase 3/metabolism , Humans , Immunohistochemistry , In Situ Nick-End Labeling , International Normalized Ratio , Ki-67 Antigen/metabolism , Liver/drug effects , Liver/pathology , Liver/physiology , Male , Organ Size , Postoperative Complications/pathology , Rats , Rats, Wistar , Recombinant Proteins , Reperfusion Injury/pathology , von Willebrand Factor/metabolism
10.
Transplantation ; 87(8): 1155-62, 2009 Apr 27.
Article in English | MEDLINE | ID: mdl-19384161

ABSTRACT

BACKGROUND AND AIM: Mitochondrial calcium overload triggers apoptosis and also regulates ATP production. ATP and uridine-5'-triphosphate (UTP) depletion from hepatic tissue after ischemia causes cell death. ATP and UTP binds to cell membranes of the hepatocytes through P2Y receptors. Our aim was to investigate the role of UTP on the hepatic injury induced by ischemia. METHODS: Isolated mouse livers were randomly divided into five groups: (1) control group; (2) ischemic group (90 min); (3) as group 2, but with the administration of UTP; (4) as group 2, but with the administration of suramin, a P2Y antagonist; and (5) as group 3, but with the simultaneous administration of suramin and UTP. RESULTS: There was a postischemic significant reduction in the release of liver enzymes in the animals pretreated with UTP, the intrahepatic caspase-3 activity was significantly decreased, and the intrahepatic ATP content increased compared with group 2 (ischemic untreated). UTP prevented intracellular Ca overload after hypoxia in hepatocyte cultures. In the UTP-treated groups, significantly fewer apoptotic hepatocyte cells were noted by weaker activation of caspase-3 and by the transferase-mediated dUTP nick end labeling assay. The administration of suramin prevented the beneficial effect of endogenous ATP. UTP treatment attenuated the degradation of IkappaBalpha (nuclear factor-kappaB inhibitor) by 80% during reperfusion with no effect on c-Jun N terminal kinase phosphorylation. CONCLUSION: The administration of UTP before induction of ischemia-reperfusion can attenuate hepatic injury. UTP administration decreased cytosolic Ca overload in hypoxic conditions. UTP-mediated protective effects may be regulated through nuclear factor- kappaB inactivation. These findings have important implications for the potential use of UTP in ischemic hepatic injury.


Subject(s)
Liver/injuries , Reperfusion Injury/prevention & control , Uridine Triphosphate/therapeutic use , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Blood Pressure , Caspase 3/metabolism , Cell Hypoxia , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/physiology , L-Lactate Dehydrogenase/metabolism , Liver/cytology , Liver/drug effects , Liver/enzymology , Male , Mice , Suramin/therapeutic use , Vena Cava, Inferior/physiology
11.
Br J Haematol ; 142(1): 82-93, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18477050

ABSTRACT

Fanconi anaemia (FA) is an autosomal recessive and X-linked disease characterized by severe genetic instability and increased incidence of cancer. One explanation for this instability may be the cellular hypersensitivity to oxidative stress leading to chromosomal breaks. This study explored the possible oxidative damage to telomeres of FA lymphocyte cell line, HSC536/N, and its possible effect on telomere function. We postulated that combination of oxidative damage with overexpression of telomerase may provide a possible model for malignant transformation in FA. The cells were grown in the presence of telomerase inhibitor and exposed for 1 month to H(2)O(2) combined with various antioxidants. This exposure caused shortening of telomere length and damage to the telomere single stranded overhang, which was prevented by several oxidants. This shortening was associated with development of severe telomere dysfunction. Control cells did not exhibit this sensitivity to H(2)O(2). Telomere dysfunction did not evoke damage response in FA cells, in contrast to normal P53 upregulation in control cells. Reconstitution of telomerase activity protected FA telomeres from further oxidative damage. These results suggest a scenario in which oxidative stress causes telomere shortening and ensuing telomere dysfunction may form the basis for malignant transformation in FA cells. Upregulation of telomerase activity in sporadic FA cells may perpetuate that process, thus explaining the malignant character of FA cells in vivo.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Fanconi Anemia/pathology , Oxidative Stress/physiology , Telomere/pathology , Anaphase/drug effects , Antioxidants/pharmacology , Cell Line , Humans , Hydrogen Peroxide/pharmacology , Oligonucleotides/pharmacology , Oxidants/pharmacology , Telomerase/antagonists & inhibitors , Telomere/drug effects , Tumor Suppressor Protein p53/metabolism
12.
Apoptosis ; 13(1): 77-86, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18085440

ABSTRACT

INTRODUCTION: Apoptosis is a central mechanism of cell death following reperfusion of the ischemic liver. Recombinant human erythropoietin (rhEPO) have an important role in the treatment of myocardial ischemia/reperfusion (I/R) injury, by preventing apoptosis. The aim of the study was to investigate the effect of different regimens of rhEPO in preventing apoptosis following I/R-induced hepatic injury. MATERIAL AND METHODS: Isolated mouse livers were randomly divided into five groups: (1) control group, perfused for the whole study period (105 min); (2) 30-min perfusion followed by 90 min of ischemia and 15 min of reperfusion; (3), (4) and (5) like group 2, but with administration of rhEPO 5,000 units/kg i.p. at 30 min, 24 h, or both 30 min and 24 h respectively, before induction of ischemia. Perfusate liver enzyme levels and intrahepatic caspase-3 activity were measured, and apoptotic cells were identified by morphological criteria, TUNEL assay, and immunohistochemistry for caspase-3. Using immunoblot the expression of the proapoptotic JNK and inhibitor of NFkappaB (IkappaBalpha) were also evaluated. von Willebrand factor (vWF) immunohistochemistry was used as a marker of endothelial cells. RESULTS: Compared to the I/R livers, all 3 rhEPO pretreated groups showed: a significant reduction in liver enzyme levels (P < 0.05) and intrahepatic caspase-3 activity (P < 0.05), fewer apoptotic hepatocytes (P < 0.05) and positive vWF staining in numerous endothelial cells lining the sinusoids. EPO decreased JNK phosphorylation and the degradation of the inhibitor of NFkappaB (IkappaBalpha) during I/R. There was no added benefit of the multiple- over the single-dose rhEPO regimen. CONCLUSION: Pretreatment with one dose of rhEPO can attenuate post-I/R hepatocyte apoptotic liver damage. NFkappaB and JNK activation is likely to play a pivotal role in the pathophysiology of I/R hepatic injury and might have a key role in EPO-mediated protective effects. This effect is associated with the increase in sinusoidal vWF immunostaining suggests an additional effect of rhEPO in liver angiogenesis recovery. These findings have important implications for the potential use of rhEPO in I/R injury during liver transplantation.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Erythropoietin/pharmacology , Liver/blood supply , Liver/pathology , MAP Kinase Kinase 4/metabolism , Protein Serine-Threonine Kinases/metabolism , Reperfusion Injury/prevention & control , Animals , Disease Models, Animal , Humans , Ischemia , Liver/metabolism , Male , Mice , Random Allocation , Recombinant Proteins , Reperfusion , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , von Willebrand Factor/metabolism , NF-kappaB-Inducing Kinase
13.
J Cell Biochem ; 104(2): 606-19, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18080320

ABSTRACT

Inflammation, elicited in the skin following tissue damage or pathogen invasion, may become chronic with deleterious consequences. Tumor necrosis factor (TNF) is a key mediator of cutaneous inflammation and the keratinocyte an important protagonist of skin immunity. Calcitriol, the hormonally active vitamin D metabolite, and its analogs attenuate epidermal inflammation and inhibit the hyperproliferation of keratinocytes associated with the inflammatory disorder, psoriasis. Since activation of extracellular signal-regulated kinase (ERK) promotes keratinocyte proliferation and mediates epidermal inflammation, we studied the effect of calcitriol on ERK activation in HaCaT keratinocytes exposed to the ubiquitous inflammatory cytokine TNF. By using the EGF receptor (EGFR) tyrosine kinase inhibitor, AG1487 and the Src family inhibitor, PP-1, we established that TNF activated ERK in an EGFR and Src dependent and an EGFR and Src independent modes. EGFR dependent activation resulted in the upregulation of the transcription factor, c-Fos, while the EGFR independent activation mode was of a shorter duration, did not affect c-Fos expression but induced IL-8 mRNA expression. Pretreatment with calcitriol, enhanced TNF-induced EGFR-Src dependent ERK activation and tyrosine phosphorylation of the EGFR, but abolished the EGFR-Src independent ERK activation. These effects were mirrored by enhancement of c-Fos and inhibition of IL-8 induction by TNF. Treatment with calcitriol increased the rate of the de-phosphorylation of activated ERK, accounting for the inhibition of EGFR-Src independent ERK activation by TNF. It is possible that effects on the ERK cascade contribute to the effects of calcitriol and its synthetic analogs on cutaneous inflammation and keratinocyte proliferation.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratinocytes/pathology , Oncogene Protein pp60(v-src)/metabolism , Tumor Necrosis Factor-alpha/physiology , Vitamin D/physiology , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cell Line , Cell Proliferation , Enzyme Activation , Humans , Inflammation/pathology , MAP Kinase Signaling System , Phosphorylation , Skin Diseases/pathology
14.
J Steroid Biochem Mol Biol ; 101(2-3): 151-60, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16889957

ABSTRACT

The anti-cancer activity of calcitriol, the active metabolite of Vitamin D, in the colon is usually attributed to its anti-proliferative and pro-differentiative actions. The levels of reactive oxygen species (ROS) are high in colon carcinomas due to increased aerobic metabolism and exposure to various anti-cancer modalities. We examined whether calcitriol modulates the response of colon cancer cells to the cytotoxic action of the common mediator of ROS injury, H2O2. Pretreatment with calcitriol (100 nM, 48 h) sensitized HT-29 colon cancer cells to cell death induced by acute exposure to H2O2 or chronic exposure to the H2O2 generating system, glucose/glucose-oxidase. Although the morphological features of H2O2-induced HT-29 cell death are consistent with apoptosis, we detected no executioner caspase activation in response to cytotoxic concentrations of H2O2 and treatment with a pan-caspase inhibitor did not affect H2O2-induced cytotoxicity nor its enhancement by calcitriol. Conversely, exposure of HT-29 cells to sub-toxic concentrations of H2O2 resulted in low executioner caspase activation that was inhibited by pretreatment with calcitriol. The sensitization of colon cancer cells to ROS-induced cytotoxicity may contribute to its assumed action as a chemopreventive agent and to its therapeutic potential alone or in combination with other anti-cancer modalities.


Subject(s)
Apoptosis , Calcitriol/pharmacology , Caspases/metabolism , Colonic Neoplasms/metabolism , Hydrogen Peroxide/pharmacology , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Synergism , HT29 Cells , Humans , Hydrogen Peroxide/metabolism , Reactive Oxygen Species/metabolism
15.
Free Radic Biol Med ; 39(2): 266-78, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15964518

ABSTRACT

Calcitriol, the hormonal form of vitamin D3, sensitizes breast cancer cells to reactive oxygen species (ROS)-dependent cytotoxicity induced by various anticancer modalities. This effect could be due to increased generation of ROS and/ or to increased sensitivity of the target cells to ROS. This work examined the effect of calcitriol on the damage inflicted on breast cancer cells by the direct action of ROS represented by H2O2. Treatment of MCF-7 cells with H2O2 resulted in activation of caspase 7 as well as induction of caspase-independent cell death. Both were enhanced by 48-72 h of pretreatment with calcitriol. This effect was not due to modulation of H2O2 degradation or to a specific effect on *OH-mediated cytotoxicity. The H2O2-induced drop in mitochondrial membrane potential and release of cytochrome c were enhanced by calcitriol. These findings indicate that calcitriol sensitizes breast cancer cells to ROS-induced death by affecting event(s) common to both caspase-dependent and -independent modes of cell death upstream to mitochondrial damage.


Subject(s)
Apoptosis , Breast Neoplasms/drug therapy , Hydrogen Peroxide/pharmacology , Vitamin D/pharmacology , Blotting, Western , Calcitriol/chemistry , Calcitriol/pharmacology , Caspases/metabolism , Cell Line, Tumor , Drug Synergism , Enzyme Activation , Humans , Hydrogen Peroxide/metabolism , Intracellular Membranes/metabolism , Membrane Potentials , Mitochondria/metabolism , Oxidants/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species , Time Factors
16.
J Steroid Biochem Mol Biol ; 89-90(1-5): 361-4, 2004 May.
Article in English | MEDLINE | ID: mdl-15225801

ABSTRACT

Calcitriol, the hormonal form of Vitamin D, potentiates the activity of some agents of the anti-cancer immune system including tumor necrosis factor-alpha (TNF-alpha). Different signaling pathways activated by TNF-alpha may be targets for calcitriol action. Activation of p38 MAP kinase was shown to have both pro- and anti-apoptotic actions in TNF-alpha-induced programmed cell death depending on cell context. Treatment of MCF-7 breast cancer cells with TNF-alpha resulted in activation of p38 MAP kinase that persisted for at least 24h. Whereas calcitriol had no effect on the earlier phase of p38 MAP kinase activation (up to 1h), it inhibited the activation of this pathway between one and 24h after exposure to TNF-alpha. Both calcitriol and the p38 MAP kinase inhibitor SB203580 enhanced TNF-alpha-induced cytotoxicity and drop in mitochondrial membrane potential, but their combined effect was sub-additive. Taken together, these findings suggest that p38 MAP kinase plays an anti-apoptotic role in TNF-alpha-induced cytotoxicity in MCF-7 cells and that the synergistic interaction between TNF-alpha and calcitriol, leading to mitochondrial damage and subsequent cell death, is partially due to modulation of this signaling pathway.


Subject(s)
Breast Neoplasms/pathology , Calcitriol/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Breast Neoplasms/enzymology , Cell Line, Tumor , Drug Synergism , Humans , Membrane Potentials , Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases
17.
Recent Results Cancer Res ; 164: 357-67, 2003.
Article in English | MEDLINE | ID: mdl-12899535

ABSTRACT

Calcitriol, the hormonal form of vitamin D, enhances the anticancer activity of the immune cytokine tumor necrosis factor, interleukin 1 and interleukin 6 in human breast and renal cell carcinoma cells without affecting the cytotoxic action of interferon-alpha or killer lymphocytes. It also enhances cytotoxicity induced by the anticancer drug doxorubicin, by the redox cycling quinone menadione, and by the reactive oxygen species hydrogen peroxide. The synergistic interaction was accompanied by increased oxidative stress, as manifested by glutathione depletion and was abolished by exposure to the thiol antioxidant N-acetylcysteine. The hormone on its own brought about an increase in the cellular redox state as reflected in the ratio between oxidized and reduced glutathione and glyceraldehyde-3-phosphate dehydrogenase, and a reduction in the expression of the antioxidant enzyme Cu/Zn superoxide dismutase. These results support the notion that the interplay between active vitamin D derivatives and other anticancer agents such as immune cytokines and anticancer drugs plays a role in the in vivo anticancer activity of vitamin D and that reactive oxygen species are involved in the anticancer activity of vitamin D on its own and in its cross-talk with other anticancer modalities.


Subject(s)
Antineoplastic Agents/pharmacology , Calcitriol/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Division/drug effects , Drug Synergism , Humans , Neoplasms/pathology
18.
Int J Cancer ; 106(2): 178-86, 2003 Aug 20.
Article in English | MEDLINE | ID: mdl-12800192

ABSTRACT

Calcitriol, the hormonal form of vitamin D, potentiates the activity of some common anticancer drugs and agents of the anticancer immune system, including tumor necrosis factor alpha (TNFalpha). TNFalpha-induced cytotoxicity is due to both caspase-dependent and -independent pathways. Cotreatment with calcitriol enhanced both modes of TNFalpha-induced death in MCF-7 breast cancer cells. It increased caspase-3-like activity as assayed by the cleavage of poly-(ADP-ribose)polymerase and of the fluorogenic substrate ac-DEVD-AMC. It also enhanced TNFalpha-induced caspase-independent cytotoxicity in the presence of the pan-caspase inhibitor zD-2,6-dichlorobenzoyloxymethylketone. The antioxidants N-acetylcysteine, reduced glutathione, lipoic acid and ascorbic acid markedly reduced the enhancing effect of the hormone on TNFalpha-induced caspase activation. N-acetylcysteine and reduced glutathione also decreased caspase-independent cytotoxicity in the presence or absence of calcitriol, indicating that reactive oxygen species (ROS) have a key role in the cross talk between TNFalpha and calcitriol. Mitochondrial damage is common to both TNFalpha-induced caspase-dependent and -independent pathways and may underlie excessive production of ROS. Mitochondrial membrane potential (DeltaPsi) was assessed by the specific potential-sensitive fluorescent probe JC-1. The hormone augmented the drop in DeltaPsi and release of cytochrome c from mitochondria, induced by TNFalpha. The effect of calcitriol on DeltaPsi was mimicked by rotenone, which increased both the drop in DeltaPsi and caspase activation induced by TNFalpha. It is possible that the interaction of TNFalpha and calcitriol on the level of the mitochondria is the underlying mechanism responsible for the enhancement of TNFalpha-induced, ROS-mediated caspase-dependent and -independent cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Calcitriol/pharmacology , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/physiology , Blotting, Western , Breast Neoplasms/metabolism , Calcium Channel Agonists/metabolism , Calcium Channel Agonists/pharmacology , Caspase 3 , Caspase Inhibitors , Cell Division , Cytochrome c Group/metabolism , Enzyme Activation , Flow Cytometry , Humans , Membrane Potentials/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2 , Rotenone/pharmacology , Signal Transduction , Tumor Cells, Cultured , Uncoupling Agents/pharmacology
19.
J Cell Biochem ; 89(3): 440-9, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12761878

ABSTRACT

The hormonally active vitamin D metabolite, 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), and keratinocyte growth factor (KGF) belong to the network of autocrine and paracrine mediators in the skin. Both were shown to modulate keratinocyte proliferation, to reverse epidermal atrophy, to increase wound healing, and to reduce chemotherapy-induced alopecia. The overlap between their activities may suggest that vitamin D exerts some of its actions by modulation of KGF activities in the skin. This notion was examined by using HaCaT keratinocytes cultured in serum-free medium in the absence of exogenous growth factors and in the presence of the EGF receptor tyrosine kinase inhibitor AG 1478 that blocks their autonomous proliferation. These cells could be stimulated to proliferate by different fibroblast growth factors (FGFs). The relative mitogenic efficacy of basic FGF, acidic FGF, or KGF was in correlation with their affinities for the KGF receptor (KGFR). Forty-eight hour co-treatment with 1,25(OH)(2)D(3) enhanced KGFR-mediated cell proliferation in a dose dependent manner. Both ERK1/2 and c-Jun N-terminal kinase (JNK) were activated by the FGFs. Treatment with 1,25(OH)(2)D(3) increased the activation of ERK but reduced the activation of JNK. Treatment with 1,25(OH)(2)D(3) increased the levels of KGFR in the presence but not in the absence of KGF, probably due to inhibition of ligand-induced receptor degradation. Inhibition of protein kinase C with bisindolylmaleimide did not interfere with the effect of 1,25(OH)(2)D(3) on KGFR-mediated ERK activation. Our results support the notion that the paracrine KGF-KGFR system in the skin can act in concert with the autocrine vitamin D system in keratinocytes to promote keratinocyte proliferation and survival under situations of stress and injury.


Subject(s)
Keratinocytes/cytology , Mitosis/drug effects , Receptors, Fibroblast Growth Factor/physiology , Vitamin D/physiology , Blotting, Western , Cell Line , Humans , Receptor, Fibroblast Growth Factor, Type 2
20.
Ann N Y Acad Sci ; 1010: 350-3, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033750

ABSTRACT

Calcitriol, the hormonal form of vitamin D, inhibited caspase-3-like activation in HaCaT keratinocytes exposed to hyperosmotic and oxidative stresses, heat shock, and the inflammatory cytokine TNF. The hormone also protected the cells from caspase-independent cell death induced by hyperosmotic and oxidative stresses. The protection against hyperosmotic stress is not affected by inhibitors of the EGF receptor, ERK or PI13 kinase pathways, neither is it due to reduced activity of the proapoptotic p38 MAP kinase. These results are in accordance with previous in vivo findings that vitamin D protects epidermal keratinocytes from apoptosis due to UV radiation or chemotherapy.


Subject(s)
Calcitriol/pharmacology , Keratinocytes/cytology , Keratinocytes/physiology , Oxidative Stress , Tumor Necrosis Factor-alpha/pharmacology , Caspase 3 , Caspase Inhibitors , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , Infant , Keratinocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...