Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 91(3): 292-5, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21763726

ABSTRACT

Although, the antiviral activity, tolerability and convenience of protease inhibitors have improved significantly in recent years, toxicity-associated adverse events including diarrhea, lipid alterations, disturbance of glucose homeostasis and liver enzyme elevations still remain a major concern during treatment of HIV-1 patients. We have recently shown that the covalent attachment of the NO moiety to the HIV-1 protease inhibitor saquinavir (Saq-NO) reduces its toxicity. In this study, we evaluated in vitro the anti-HIV activity of Saq-NO vs. its parental compound Saq. Site directed mutants with the most frequently identified Saq associated resistance mutations and their combinations were generated on proviral AD8-based backbones. Phenotypic assays were conducted using wild type clinical isolates and fully replicating recombinant viruses with Saq and Saq-NO in parallel on purified CD4+ T cells. The following recombinant viruses were generated and tested: L33F, M46I, G48V, I54V, I84V + L90M, M46I + L90M, G48V + L90M, M46I + I54V + L90M, L33F + M46I + L90M. The fold change resistance compared to the wild type viruses was between 1.3 and 7 for all single mutants, between 3.4 and 20 for double mutants and between 16.7 and 28.5 for viruses carrying three mutations for both compounds. The results clearly demonstrate that Saq-NO maintains an anti-HIV-1 profile very similar to that of Saq. The possibility to reduce Saq associated side effects and to increase the concentration of the drug in vivo may allow a higher and possibly more effective dosage of Saq-NO in HIV-1-infected patients and to increase the genetic barrier of this PI thus impairing the selection of resistant clones.


Subject(s)
Drug Resistance, Viral/drug effects , HIV Infections/drug therapy , HIV-1/drug effects , Saquinavir/analogs & derivatives , Saquinavir/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Survival/drug effects , DNA Primers , HIV Infections/virology , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , HIV-1/enzymology , HIV-1/genetics , Humans , Inhibitory Concentration 50 , Mutagenesis, Site-Directed , Saquinavir/chemistry , Saquinavir/therapeutic use
2.
Mol Cancer Ther ; 8(5): 1169-78, 2009 May.
Article in English | MEDLINE | ID: mdl-19417156

ABSTRACT

Application of the HIV protease inhibitor saquinavir (Saq) to cancer chemotherapy is limited by its numerous side effects. To overcome this toxicity, we modified the original compound by covalently attaching a nitric oxide (NO) group. We compared the efficacy of the parental and NO-modified drugs in vitro and in vivo. The novel compound saquinavir-NO (Saq-NO) significantly reduced the viability of a wide spectrum of human and rodent tumor cell lines at significantly lower concentration than the unmodified drug. In contrast to Saq, Saq-NO had no effect on the viability of primary cells and drastically reduced B16 melanoma growth in syngeneic C57BL/6 mice. In addition, at the equivalent of the 100% lethal dose of Saq, Saq-NO treatment caused no apparent signs of toxicity. Saq-NO blocked the proliferation of C6 and B16 cells, up-regulated p53 expression, and promoted the differentiation of these two cell types into oligodendrocytes or Schwann-like cells, respectively. Although it has been well documented that Saq decreases tumor cell viability by inhibiting Akt, the anticancer properties of Saq-NO were completely independent of the phosphatidylinositol 3-kinase/Akt signaling pathway. Moreover, Saq-NO transiently up-regulated Akt phosphorylation, delivering a protective signal that could be relevant for primary cell protection and the absence of drug toxicity in vivo. It was unlikely that released NO was independently responsible for these drug effects because Saq-NO treatment increased intracellular and secreted NO levels only slightly. Rather, the chemical modification seems to have produced a qualitatively new chemical entity, which may have a unique mode of action against cancer cells.


Subject(s)
Antineoplastic Agents , Neoplasms/metabolism , Nitric Oxide , Oncogene Protein v-akt/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Saquinavir , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytostatic Agents/pharmacology , Drug Synergism , Humans , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Rats , Saquinavir/chemistry , Saquinavir/pharmacology , Saquinavir/toxicity , Signal Transduction/drug effects
3.
BMC Biotechnol ; 8: 85, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19014469

ABSTRACT

BACKGROUND: Human monoclonal antibodies (mAbs) generated as a result of the immune response are likely to be the most effective therapeutic antibodies, particularly in the case of infectious diseases against which the immune response is protective.Human cytomegalovirus (HCMV) is an ubiquitous opportunistic virus that is the most serious pathogenic agent in transplant patients. The available therapeutic armamentarium (e.g. HCMV hyperimmune globulins or antivirals) is associated with severe side effects and the emergence of drug-resistant strains; therefore, neutralizing human mAb may be a decisive alternative in the prevention of primary and re-activated HCMV infections in these patients. RESULTS: The purpose of this study was to generate neutralizing mAb against HCMV from the immunological repertoire of immune donors. To this aim, we designed an efficient technology relying on two discrete and sequential steps: first, human B-lymphocytes are stimulated with TLR9-agonists and IL-2; second, after both additives are removed, the cells are infected with EBV. Using this strategy we obtained 29 clones secreting IgG neutralizing the HCMV infectivity; four among these were further characterized. All of the mAbs neutralize the infection in different combinations of HCMV strains and target cells, with a potency approximately 20 fold higher than that of the HCMV hyperimmune globulins, currently used in transplant recipients. Recombinant human monoclonal IgG1 suitable as a prophylactic or therapeutic tool in clinical applications has been generated. CONCLUSION: The technology described has proven to be more reproducible, efficient and rapid than previously reported techniques, and can be adopted at low overall costs by any cell biology laboratory for the development of fully human mAbs for immunotherapeutic uses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Cytomegalovirus Infections/therapy , Cytomegalovirus/immunology , Amino Acid Sequence , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/biosynthesis , Antibodies, Viral/therapeutic use , B-Lymphocytes/virology , Base Sequence , Cell Line , Cells, Cultured , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Humans , Interleukin-2/metabolism , Molecular Sequence Data , Neutralization Tests , Receptors, IgE/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Reproducibility of Results , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/metabolism , Viral Envelope Proteins/metabolism
4.
Cytokine ; 42(3): 372-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18455423

ABSTRACT

Major hepatic resection in cirrhotic patients is associated with impaired liver regeneration and failure, leading to high peri-operative mortality. In this work, the causes of defective regeneration in cirrhotic liver and the utility of IL-6 treatment were investigated in an experimental model combining cirrhosis and partial hepatectomy in the rat. Relative to normal controls, decompensated cirrhotic animals showed decreased survival, while compensated cirrhotic animals showed similar survival but reduced hepatic DNA synthesis and newly regenerated liver mass amount. Defective liver regeneration was associated with a decrease in STAT3 and NF-kB activation, consistent with an increased accumulation of their respective inhibitors PIAS3 and IkBalpha, and with a decreased induction of Bcl-xL. Treatment with recombinant IL-6 enhanced survival of decompensated cirrhotic animals, while it did not affect survival of compensated cirrhotic animals but sustained liver regeneration, by restoring STAT3 and NF-kB activation and Bcl-xL induction to the levels found in normal controls. The pro-growth effects exerted by IL-6 treatment in cirrhotic liver were attained also at low, pharmacologically acceptable doses. In conclusion, our results suggest that IL-6 treatment may be therapeutic in major resection of cirrhotic liver.


Subject(s)
Interleukin-6/pharmacology , Liver Cirrhosis, Experimental/physiopathology , Liver Regeneration/drug effects , Recombinant Proteins/pharmacology , Animals , Hepatectomy , Hepatocytes/physiology , Humans , I-kappa B Proteins/metabolism , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/surgery , Male , Molecular Chaperones/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Protein Inhibitors of Activated STAT/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , bcl-X Protein/metabolism
5.
Nitric Oxide ; 19(2): 177-83, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18460348

ABSTRACT

In this study we evaluated the effects of the new NO donating compound (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid-nitric oxide (GIT-27NO) on the A375 human melanoma cell line. Treatment with the drug led to concentration-dependent reduction of mitochondrial respiration and number of viable cells in cultures. Decreased cell viability correlated with release and internalization of NO and was neutralized by the extracellular scavenger hemoglobin. GIT-27NO neither influenced cell division nor induced accidental or autophagic cell death. Early signs of apoptosis were observed upon coculture with the drug, and resulting in marked accumulation of hypodiploid cells, suggesting that the induction of apoptosis is one primary mode of action of the compound in A375 cells. GIT-27NO significantly inhibited the expression of the transcription repressor and apoptotic resistant factor YY1 and, in parallel, augmented the presence of total p53. The capacity of GIT-27NO to induce p53-mediated apoptosis along with inhibition of YY1 repressor in A375 melanoma cells indicates that GIT-27NO possesses an important anti-cancer pharmacological profile. The findings suggest the potential therapeutic use of GIT-27NO in the clinical setting.


Subject(s)
Apoptosis/drug effects , Melanoma/drug therapy , Nitric Oxide Donors/pharmacology , Tumor Suppressor Protein p53/drug effects , Acetates , Antineoplastic Agents , Cell Line, Tumor , Cell Survival/drug effects , Humans , Melanoma/pathology , Nitric Oxide Donors/therapeutic use , Oxazoles , YY1 Transcription Factor/antagonists & inhibitors
6.
Mol Cancer Ther ; 7(3): 510-20, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18347138

ABSTRACT

Preclinical studies have shown that nitric oxide (NO)-donating nonsteroidal anti-inflammatory drugs possess anticancer activities. Here, we report in vitro and in vivo studies showing the antitumor effect of the NO-donating isoxazole derivative (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid (GIT-27NO). GIT-27NO, but not the NO-deprived parental compound VGX-1027, significantly affected viability of both rodent (L929, B16, and C6) and human (U251, BT20, HeLa, and LS174) tumor cell lines. GIT-27NO triggered either apoptotic cell death (e.g., L929 cells) or autophagic cell death (C6 and B16 cells). Moreover, GIT-27NO hampered the viability of cisplatin-resistant B16 cells. NO scavenger hemoglobin completely prevented GIT-27NO-induced death, indicating that NO release mediated the tumoricidal effect of the compound. Increase in intracellular NO upon on the treatment was associated with intensified production of reactive oxygen species, whereas their neutralization by antioxidant N-acetylcysteine resulted in partial recovery of cell viability. The antitumor activity of the drug was mediated by the selective activation of mitogen-activated protein kinases in a cell-specific manner and was neutralized by their specific inhibitors. In vivo treatment with GIT-27NO significantly reduced the B16 melanoma growth in syngeneic C57BL/6 mice. The therapeutic effect occurred at dose (0.5 mg/mouse) up to 160 times lower than those needed to induce acute lethality (80 mg/mouse). In addition, a dose of GIT-27NO five times higher than that found effective in the melanoma model was well tolerated by the mice when administered for 4 consecutive weeks. These data warrant additional studies to evaluate the possible translation of these findings to the clinical setting.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Nitric Oxide Donors/pharmacology , Oxazoles/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Nitric Oxide Donors/toxicity
7.
Hepatogastroenterology ; 54(75): 878-83, 2007.
Article in English | MEDLINE | ID: mdl-17591083

ABSTRACT

BACKGROUND/AIMS: In the liver IL-6 displays growth-inducing and pro-survival activities. We studied the pro-proliferative and protective mechanisms of IL-6 treatment in a model of liver cirrhosis in wild type rat, investigating the theoretical basis for a potential pharmacologic role of IL-6 in cirrhosis. METHODOLOGY: We analyzed IL-6 receptors levels in cirrhotic liver. We also studied the activation of signaling pathways downstream IL-6 receptors by analyzing the DNA-binding activity of transcription factors STAT3, AP-1, HNF-1 and NF-kappaB and the phosphorylation status of AKT and eNOS. We also analyzed hepato-cell proliferation, by determining BrdU incorporation into DNA, and liver mass expansion. RESULTS: We show that liver cells from cirrhotic animals have increased expression of the IL-6 receptor alpha/gp80. In addition, we show that in cirrhosis the main molecular pathways downstream the receptors are intact and that IL-6 activates STAT3, AP-1 and NF-kappaB transcription factors, induces AKT and eNOS phosphorylation and increases hepato-cell proliferation and liver mass expansion in a dose-dependent manner. CONCLUSIONS: Our data demonstrate that the theoretical basis exists for the therapeutic employment of IL-6 in liver cirrhosis.


Subject(s)
Interleukin-6/pharmacology , Liver Cirrhosis/metabolism , Liver Regeneration , Liver/drug effects , Receptors, Interleukin-6/metabolism , Animals , Bromodeoxyuridine/analysis , Cytokine Receptor gp130/metabolism , Liver/metabolism , Male , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism
8.
Cytokine ; 34(3-4): 131-42, 2006 May.
Article in English | MEDLINE | ID: mdl-16814559

ABSTRACT

Numerous animal studies simulating liver injury have demonstrated that interleukin-6 (IL-6) exerts a protective effect. This study was designed to further analyze the molecular mechanisms underlying the protective role of IL-6 in a rat model of liver ischemia/reperfusion injury. We show that IL-6: (i) at high doses reduces cell damage which occurs in ischemic-reperfused liver, while at low doses displays only a limited protective capacity, (ii) anticipates and enhances hepatocyte compensatory proliferation seen in ischemic-reperfused liver also at a low, more pharmacologically acceptable dose, (iii) sustains the acute phase response which is dampened in ischemic-reperfused liver, (iv) strengthens the heat shock-stress response shown by ischemic-reperfused liver and (v) overcomes the dysfunctions of the unfolding protein response found in ischemic-reperfused liver. We also show that IL-6-enhanced STAT3 activation probably plays a crucial role in the potentiation of the different protective pathways activated in ischemic-reperfused liver. Our data confirm that IL-6 is a potential therapeutic in liver injury of different etiologies and reveal novel mechanisms by which IL-6 sustains liver function after ischemia/reperfusion injury.


Subject(s)
Interleukin-6/pharmacology , Liver/drug effects , Reperfusion Injury/prevention & control , Acute-Phase Reaction , Animals , DNA/biosynthesis , Disease Models, Animal , Gene Expression Regulation/drug effects , Heat-Shock Response/drug effects , Liver/cytology , Liver/pathology , Protein Denaturation/drug effects , Rats , Rats, Wistar , STAT3 Transcription Factor/metabolism
9.
Neuropathology ; 26(1): 32-42, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16521477

ABSTRACT

Neuropathy is the most severe and the least understood complication of diabetes. We investigated the potential neuroprotective effect of IL-6 therapy in an experimental model of diabetic neuropathy. A single i.v. injection of streptozotocin (STZ, 55 mg/kg) was used to induce experimental diabetes in adult males. IL-6 (1, 10 or 30 microg/kg) was administrated either intraperitoneally on a daily basis or subcutaneously (s.c.) on a daily, on a three times or one time per week basis, starting at day 10 post-STZ. A decrease in sensory nerve conduction velocity (SNCV), indicative of neuropathy, is seen in STZ rats as early as day 10 post-STZ, a time at which blood glycaemia is already maximal. At later time points, this electrophysiological impairment became severe and clinically apparent by affecting tail flick latency. Motor dysfunction defined by a significant increase in compound muscle action potential (CMAP) latency was also recorded. At the completion of the study (day 40 post-STZ), histological examination revealed significant axonopathy and myelin loss, along with an increase in the proportion of fibers with abnormal appearance in sciatic nerves of STZ rats. These changes were not observed in non-diabetic rats and were significantly prevented by IL-6 treatment. The optimal dose appeared to be 10 microg/kg s.c. three injections per week, which showed a better effect in most of the parameters studied than 4-methylcatechol, a NGF-like neuroprotective compound. Once weekly and three times weekly administrations of IL-6 were as effective as daily treatment. Taken together, these results support the potential neuroprotective actions of IL-6. The fact that the half-life of IL-6 is only approximately 5 h while weekly dosing was neuroprotective strongly suggests activation by IL-6 of effector molecule(s) with longer duration of action.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/prevention & control , Interleukin-6/therapeutic use , Action Potentials/drug effects , Animals , Catechols/therapeutic use , Diabetic Neuropathies/pathology , Electromyography , Male , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley
10.
Cytokine ; 30(5): 236-42, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15927847

ABSTRACT

Interleukin-6 (IL-6) reduces myocardial haemodynamics. However, the intrinsic mechanisms of IL-6 effects are not known. We hypothesized that nitric oxide (NO) synthesised by neuronal synthase (nNOS) can be the molecular mediator of IL-6-mediated cardiac effects. Thus, we investigated in vivo after IL-6 acute administration: (1) the role of NO pathway; (2) the importance of NO derived from nNOS located in intracardiac vagal ganglion in the anterior surface of the left ventricle. Sprague-Dawley (SD) rats (225-250 g) were anaesthetized (sodium pentobarbital 30 mg/kg intraperitoneally administered) and ventilated. The effects of a single IL-6 bolus (100 microg/kg intravenously administered) were studied in four experimental groups: (a) IL-6 (n=6), (b) IL-6 plus 30 mg/kg of L-NAME (an eNOS and nNOS inhibitor; n=6), (c) IL-6 plus 25mg/kg of 7-NI (a specific nNOS inhibitor; n=6), (d) IL-6 plus vagal resection (n=6). We evaluated the following parameters: mean aortic pressure (MAP), left ventricular end systolic pressure (LVESP), left ventricular positive peak dP/dt (PP dP/dt). Data are expressed as mean+/-sem. IL-6 caused a transient but significant reduction of MAP (-21.8% of basal: p<0.05), LVESP (from 130+/-4.2 to 1056.5 mmHg: p<0.05) and PP dP/dt (from 5390+/-158 to 4400+/-223 mmHg/s, p<0.02). Concomitant treatment with L-NAME or 7-NI totally abolished IL-6 effects. Vagal resection significantly reduced the haemodynamic effects (MAP: -10% of basal: p=ns; LVEDS: from 125+/-7.3 to 117+/-6.8 mmHg, p<0.05; PP dP/dt from 5500+/-150 to 5000+/-143 mmHg/s, p<0.05). We conclude that acute administration of IL-6 caused transient but significant cardiac negative inotropism. IL-6 haemodynamic effects are partly due to NO synthesised by nNOS located in vagal left ventricular ganglia.


Subject(s)
Heart/drug effects , Heart/physiology , Interleukin-6/pharmacology , Nitric Oxide/blood , Vagus Nerve/drug effects , Vagus Nerve/physiology , Animals , Blood Pressure/drug effects , Blotting, Western , Enzyme Inhibitors/pharmacology , Heart/innervation , Hemodynamics/drug effects , In Vitro Techniques , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
11.
J Med Chem ; 47(14): 3546-60, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15214782

ABSTRACT

New 5alpha-reductase 1 (5alphaR-1) inhibitors were designed to complete a consistent set of analogues suitable for a 3D QSAR study. These compounds were synthesized by a modification of the aza-Robinson annulation, further functionalized by Pd-catalyzed cross-coupling processes, and were tested with human 5alphaR-1 expressed in Chinese hamster ovary 1827 cells. It turned out that the potency of the resulting inhibitors was strongly dependent on the type of substitution at the 8 position, with the IC(50) values ranging from 8.1 to 1050 nM. The construction of this homogeneous set of molecules allowed a 3D QSAR study. In particular, comparative molecular field analysis (CoMFA) was used to correlate the potency of the inhibitors with their physicochemical features. Highly accurate evaluations of the atomic point charges were carried out by means of quantum chemical calculations at the DFT/B3LYP level of theory followed by the RESP fitting procedure. It turned out that increasing the reliability of electrostatic parameters greatly affected the statistical results of the QSAR analysis. The 3D QSAR model proposed could be very useful in the further development of 5alphaR-1 inhibitors, which are suitable candidates to be evaluated as drugs in the treatment of 5alphaR-1 related diseases such as acne and alopecia in men and hirsutism in women.


Subject(s)
5-alpha Reductase Inhibitors , Quinolizines/chemical synthesis , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/chemistry , Animals , CHO Cells , Cricetinae , Humans , Models, Molecular , Molecular Conformation , Quantitative Structure-Activity Relationship , Quinolizines/chemistry , Quinolizines/pharmacology
12.
Exp Hematol ; 32(2): 171-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15102478

ABSTRACT

OBJECTIVE: The aim of this story was to evaluate the activity of recombinant human (rh) growth hormone (GH) in restoring bone marrow progenitor cell growth as well as cytokine-elicited stem cell mobilization in aged BALB/c mice with impaired marrow hematopoietic function and reduced stem cell mobilizing capacity. MATERIALS AND METHODS: BALB/c mice included in this study were either naturally aged (group I) or aged after having been used for radioprotective assays (group II). Mice were treated for 5 weeks with either rhGH [2.5 mg/kg/day intraperitoneally (IP)] or phosphate-buffered saline (PBS). Subsequently, colony-forming cells (CFCs) and long-term culture-initiating cells (LTC-ICs) were evaluated. In addition, progenitor cell mobilization elicited by granulocyte colony-stimulating factor (rhG-CSF) was analyzed. RESULTS: Compared with young controls, the growth of marrow CFCs and LTC-ICs was significantly reduced (P < or = 0.05) in group I and II mice. Treatment with rhGH significantly enhanced marrow hematopoiesis in mice of both groups, as demonstrated by a complete restoration of marrow cellularity, and CFC and LTC-IC growth. To further evaluate the hematopoietic potential of rhGH, aged mice treated with rhGH or PBS were mobilized with rhG-CSF (10 microg/day IP for 5 days). Compared with PBS-injected mice, rhGH-treated mice showed a significant improvement of rhG/CSF-elicited stem cell mobilization, with significant increases of white blood cell counts (5633 vs 8133, P < or = 0.05), frequency of circulating CFCs per 10(5) mononuclear cells (36 vs 67, P < or = 0.009), as well as absolute numbers per mL of blood of circulating CFCs (783 vs 2288, P < or = 0.001) and LTC-IC (21 vs 64, P < or = 0.001). CONCLUSION: Our data demonstrate in mice that a 5-week treatment with rhGH restores age- and irradiation-associated loss of marrow primitive and committed progenitors.


Subject(s)
Bone Marrow Cells/radiation effects , Growth Hormone/pharmacology , Hematopoietic Stem Cells/radiation effects , Age Factors , Animals , Bone Marrow Cells/drug effects , Female , Hematopoiesis/drug effects , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Mice , Mice, Inbred BALB C
13.
J Virol ; 78(6): 3190-5, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990741

ABSTRACT

Human cytomegalovirus (HCMV) exploits the host transcription factor NF-kappaB to enhance its own replication, dissemination, and reactivation from latency. Here we report that HCMV infection activates the upstream IkappaB kinase (IKK) complex and that its catalytic IKK2 subunit is required for HCMV-induced NF-kappaB activation, as well as the replication of different HCMV strains. These results indicate that IKK2 is essential for HCMV replication and emphasize the feasibility of blocking NF-kappaB activation as a way of inhibiting infection.


Subject(s)
Cytomegalovirus/pathogenicity , Protein Serine-Threonine Kinases/metabolism , Virus Replication , Cell Line , Cytomegalovirus/physiology , Gene Expression Regulation , Humans , I-kappa B Kinase , NF-kappa B/metabolism , Transcriptional Activation
14.
Mol Cell Neurosci ; 25(2): 301-11, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15019946

ABSTRACT

We investigated the effects of IL-6 and a chimeric derivative of IL-6 and soluble IL-6 receptor (IL6RIL6 chimera) on excitotoxic injury in rat organotypic hippocampal slices. Brief application of N-methyl-d-aspartate (NMDA) induced astrocyte reactivity, neuron cell death, and oligodendrocyte degeneration, the latter caused by secondary activation of AMPA/kainate receptors. Both these cytokines rescued neurons and oligodendrocytes, albeit the chimeric compound was much more potent and efficient than IL-6. No change was produced on reactive astrocytosis. The cytokines preserved myelin basic protein (MBP) production in slices exposed to excitotoxic insult, and when applied singularly for a week, they also enhanced both MBP and proteolipid protein expression. These effects occurred through activating the signal transducer gp130 and were associated with stimulation of transcription factors STAT1 and STAT3. Our results suggest that IL-6 and IL6RIL6 may prove to be valuable in treating neurodegenerative and demyelinating diseases.


Subject(s)
Interleukin-6/pharmacology , Nerve Degeneration/prevention & control , Neurons/drug effects , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Animals , Antigens, CD/metabolism , Astrocytes/drug effects , Astrocytes/physiology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cytokine Receptor gp130 , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Gliosis/physiopathology , Gliosis/prevention & control , Hippocampus/cytology , In Vitro Techniques , Interleukin-6/genetics , Membrane Glycoproteins/metabolism , Myelin Basic Protein/genetics , Myelin Proteolipid Protein/genetics , N-Methylaspartate/antagonists & inhibitors , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Neurons/metabolism , Neurotoxins/antagonists & inhibitors , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Phosphorylation/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Receptors, Interleukin-6/genetics , Recombinant Fusion Proteins/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , Trans-Activators/drug effects , Trans-Activators/metabolism
15.
J Clin Endocrinol Metab ; 88(11): 5537-46, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14602802

ABSTRACT

LH and human chorionic gonadotropin (hCG) control steroid production and gametogenesis. They also function as growth factors through interaction with a specific receptor that is a member of the seven-transmembrane receptor family coupled via G proteins to signal pathways involving cAMP and phospholipase C/inositol 3 phosphate. For this study, monoclonal antibodies (mAbs) were raised against the human LH receptor (LHR)/hCG receptor (hCGR), using Chinese hamster ovary LHR-transfected cells as the immunogen. Two reagents were then selected on the basis of their ability to recognize the full-length transmembrane receptor expressed both by Chinese hamster ovary LHR-transfected cells and by a limited number of tumor cell lines. One of these mAbs reacts with the LHR/hCGR in tissue sections of both frozen and paraffin-embedded specimens. This unique feature allowed us to map the cytological distribution of LHR/hCGR in human breast tissues at different stages of development in physiological and benign pathological conditions. The same mAb proved to be agonistic: receptor ligation elicits signals that modulate the growth of selected breast tumor cell lines. This observation suggests that the mAb recognizes an epitope that is included in the domain of the receptor involved in the interaction with the natural ligand.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptors, LH/immunology , Receptors, LH/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Breast/chemistry , Breast/metabolism , CHO Cells , Cell Division/immunology , Cell Line, Tumor , Cricetinae , Cyclic AMP/metabolism , Epitope Mapping , Female , Fixatives , Formaldehyde , Hybridomas , Immunohistochemistry , Mice , Mice, Inbred BALB C , Paraffin Embedding , Receptors, LH/analysis
16.
Mol Cell Neurosci ; 21(4): 602-15, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12504593

ABSTRACT

We investigated the effects of a chimeric protein (IL6RIL6 chimera) containing interleukin-6 (IL-6) fused to its soluble receptor (sIL-6R) on the proliferation and/or differentiation of rat oligodendrocyte progenitor cells (OPCs) and on oligodendrocyte survival. Exposure of OPCs to IL6RIL6 chimera for 48 h induced a dose-dependent decrease of bromodeoxyuridine (BrdU) incorporation. IL6RIL6 chimera treatment for 48 h also strongly increased the reduction of the tetrazolium salt 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) by mitochondrial enzymes and enhanced oligodendrocyte staining with a mitochondrial fluorescent dye. A strong, dose-dependent increase in the number and length of processes immunostained for early (galactocerebroside) or late (myelin basic protein) oligodendrocyte differentiation markers was revealed after OPC treatment with IL6RIL6 chimera for 2-7 days, respectively. Moreover, treatment with IL6RIL6 chimera improved oligodendrocyte survival. The chimera-induced increase of oligodendrocyte arborization was mimicked, although with lower efficacy, by ciliary neurotrophic factor (CNTF) but not by IL-6 and was reduced in the presence of a gp130 soluble peptide which is able to inhibit the gp130-mediated signals of the IL-6/sIL-6R complex. Oligodendrocyte treatment with IL6RIL6 chimera for 30 min induced both signal transducer and the activator of transcription-1 (STAT-1) and STAT-3 phosphorylation and nuclear translocation. We conclude that, by interacting with membrane gp130 and possibly by activating Janus kinase/STAT pathways, IL6RIL6 chimera induces OPCs to differentiate into mature oligodendrocytes, promotes their survival, and could deserve investigation as a therapeutic strategy for enhancing remyelination.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Interleukin-6/immunology , Oligodendroglia/drug effects , Receptors, Interleukin-6/immunology , Recombinant Fusion Proteins/pharmacology , Stem Cells/drug effects , Animals , Animals, Newborn , Cell Differentiation/immunology , Cell Division/drug effects , Cell Division/genetics , Cell Division/immunology , Cell Lineage/immunology , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Ciliary Neurotrophic Factor/immunology , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Growth Substances/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oligodendroglia/immunology , Oligodendroglia/metabolism , Rats , Receptors, Cytokine/immunology , Receptors, Cytokine/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , STAT1 Transcription Factor , STAT3 Transcription Factor , Stem Cells/immunology , Stem Cells/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...