Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Regul Toxicol Pharmacol ; 72(2): 169-78, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25896096

ABSTRACT

Emricasan, formerly IDN-6556, is a small molecule currently being evaluated in clinical trials to reduce hepatic injury and liver fibrosis. Since emricasan is an irreversible pan-caspase inhibitor that potently inhibits caspase-mediated apoptosis and inflammation, its carcinogenic potential was evaluated in a humanized mouse model. Tg.rasH2 mice received LabDiet formulated with 0, 10, 25, and 75mg/kg/day of emricasan, for 26weeks. At terminal sacrifice, blood was collected for clinical pathology analysis and tissues were collected, processed, and evaluated microscopically. There were no treatment related deaths or overt signs of toxicity for the duration of the study. There was no evidence of a carcinogenic effect in the peripheral blood leukocyte counts. Liver microgranulomas, which are background lesions, were slightly increased, especially in males. Increases in the incidence of the activated germinal centers were seen in the spleens and mesenteric lymph nodes of males and females, and in the mandibular lymph nodes of male mice. Atrophy of ovaries and testicular degeneration were also seen in emricasan treated animals. Although several non-neoplastic lesions were observed, there was no evidence of emricasan-related tumor formation in any tissue. In addition, the non-neoplastic lesions were not considered pre-neoplastic. Thus, emricasan is not considered carcinogenic.


Subject(s)
Caspase Inhibitors/toxicity , Pentanoic Acids/toxicity , Animals , Carcinogenicity Tests , Caspase Inhibitors/blood , Caspase Inhibitors/pharmacokinetics , Female , Genes, ras , Granuloma/chemically induced , Liver/drug effects , Liver/pathology , Lung/anatomy & histology , Lung/drug effects , Male , Mice, Transgenic , Ovary/drug effects , Ovary/pathology , Pentanoic Acids/blood , Pentanoic Acids/pharmacokinetics , Spleen/anatomy & histology , Spleen/drug effects , Testis/drug effects , Testis/pathology
2.
Chemotherapy ; 52(6): 308-15, 2006.
Article in English | MEDLINE | ID: mdl-17008791

ABSTRACT

BACKGROUND: Chemotherapy is effective against a wide variety of tumor cells, although its use is limited by side effects. In vitro experiments and phase I and II trials have shown that phytochemicals such as perillyl alcohol (P-OH) have antitumor effects. Pentoxifylline (PTX), a synthetic methylxanthine used mainly to treat pathologies associated with hematological diseases, sensitizes tumor cells to chemotherapy. The aim of this study was to determine whether PTX amplifies the antitumor effects of P-OH in U937 human myelomonocytic leukemia cells. METHODS: Apoptosis was measured by the loss of mitochondrial membrane potential determined by flow cytometry using dihexyloxacarbocyanine iodide (DiOC6) and propidium iodide. Bcl-2 and Bax protein expression was also assessed by Western blot analysis. RESULTS: P-OH and PTX induced loss of the mitochondrial membrane potential in U937 cells in vitro. Culturing the cells in the presence of both compounds caused a significant increase (p < 0.001) in apoptosis and expression of anti-apoptotic Bcl-2 and pro-apoptotic Bax proteins. However, despite their coexistence, Bax expression prevailed in our experiments. These data suggest that the effects of PTX might be attributable to changes in the mitochondrial membrane potential. CONCLUSION: PTX sensitizes tumor cells to the anti-neoplastic action of P-OH. These observations may have clinical relevance in the treatment of cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Monoterpenes/pharmacology , Pentoxifylline/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/metabolism , Blotting, Western , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Flow Cytometry , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/physiopathology , Membrane Potential, Mitochondrial/drug effects , Monoterpenes/administration & dosage , Pentoxifylline/administration & dosage , Tumor Cells, Cultured , U937 Cells
3.
Cancer Lett ; 229(1): 59-65, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16157219

ABSTRACT

Apoptosis was followed in L5178Y lymphoma cell-bearing mice at different times after intraperitoneal injections of adriamycin (ADM). Apoptosis was determined morphologically and confirmed by DNA laddering on electrophoresis. Apoptosis was observed 36h after injection of 5mg/kg ADM (apoptotic cell index 64.2+/-5.6 vs. 1.5+/-2.1 from the untreated group) and confirmed by DNA electrophoresis. However, when the animals were pretreated with (+)-alpha-tocopherol acid succinate or superoxide dismutase before ADM administration apoptotic index significantly diminished (P<0.05) and the DNA electrophoresis did not show fragmentations. We conclude that in ADM-treated mice, tumour cell death occurs in two ways: first by necrosis, then later by apoptosis. These observations are likely to be associated with or caused by the generation of reactive oxygen species.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Doxorubicin/pharmacology , Lymphoma/pathology , Superoxide Dismutase/metabolism , Vitamin E/analogs & derivatives , Animals , Injections, Intraperitoneal , Lymphoma/veterinary , Male , Mice , Mice, Inbred BALB C , Necrosis , Reactive Oxygen Species/adverse effects , Tocopherols , Transplantation, Heterologous , Vitamin E/pharmacology
4.
Anticancer Res ; 21(3B): 1869-72, 2001.
Article in English | MEDLINE | ID: mdl-11497270

ABSTRACT

Adriamycin (ADM) is an oncostatic of the anthracycline family with confirmed experimental and clinical efficiency. This antitumoral drug has been reported to stimulate macrophage activity and is able to induce apoptosis (AP) in some tumour cells. The objective of the present work was to investigate if in vivo administration of ADM to mice induces AP in their peritoneal macrophages (PM). AP was expressed by the apoptotic index (AI) of peritoneal macrophages observed under fluorescence microscope after ethidium bromide and acridine orange staining and confirmed by detection of the ladder pattern on DNA electrophoresis, indicates DNA fragmentation in 80-120 bp characteristic of apoptotic state. 24 hours after i.p. ADM administration, AP was observed in PM. The effect was best visible after the injection of 5 mg/kg ADM. (Al: 76.3+/-8.9 vs untreated control group AI: 2.8+/-1.1). In the ADM treated group a DNA ladder electrophoretic pattern was observed while DNA from normal PM was genomic. Since ADM toxicity has been attributed to reactive oxygen species generation, we investigated its possible participation in AP induction by pretreating mice with antioxidants: (+)-alpha-tocopherol acid succinate (30 IU/mouse per os) for 3 days before ADM administration with E. coli lipopolysacharide (0.15 microg/mouse i.p.) 24 hours before ADM administration or with superoxide dismutase (10,000 IU/mouse i.p.) 1 hour before ADM administration. AI was significantly decreased, with values close to those of the untreated control group (AI: 15+/-5.7, 9.6+/-8.0 and 32.9+/-6.9, respectively). Antioxidants given before ADM treatment significantly increased the live cell index (p < or = 0.001) in PM the groups while inactivated antioxidants no longer protect PM against the ADM AP induction. DNA analysis confirmed the effect: in the untreated control and in the antioxidant protected groups DNA was genomic while in either ADM or inactivated-antioxidants + ADM treated groups, DNA presented the ladder pattern. AP can thus be induced in PM by ADM and inhibited by antioxidants. These observations may have clinical applications.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis , Doxorubicin/pharmacology , Macrophages, Peritoneal/pathology , Vitamin E/analogs & derivatives , Acridine Orange/pharmacology , Animals , Cell Survival , DNA Fragmentation , Ethidium/pharmacology , Fluorescent Dyes/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Reactive Oxygen Species , Superoxide Dismutase/pharmacology , Tocopherols , Vitamin E/pharmacology
5.
J Pharmacol Exp Ther ; 297(2): 811-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11303074

ABSTRACT

Previous studies have shown that caspase inhibitors are effective at protecting against anti-Fas antibody (alpha-Fas)-mediated liver injury/lethality. The purpose of these experiments was to characterize more fully the efficacy of a broad-spectrum, irreversible caspase inhibitor, IDN-1965 (N-[(1,3-dimethylindole-2-carbonyl)valinyl]-3-amino-4-oxo-5-fluoropentanoic acid), in this model and the role of caspase inhibition in long-term protection. The ED(50) for IDN-1965 by i.p. administration, based on alanine aminotransferase activities, was 0.14 mg/kg. The caspase inhibitor was also efficacious when administered intravenously and orally (ED(50) values of 0.04 and 1.2 mg/kg, respectively). Histologically, marked reduction in Fas-induced apoptosis with IDN-1965 (1 mg/kg, i.p.) was apparent at 6 h. Also, caspase 3-like activities were decreased in a dose-dependent manner, but the inhibition of caspase activity was transient. Immunohistochemical studies demonstrated that IDN-1965 greatly reduced the activation of caspase 3. In survival studies, a single i.p. treatment of 1 mg/kg IDN-1965 or continuous i.p. infusion via osmotic pumps completely blocked lethality measured up to 7 days after alpha-Fas administration. IDN-1965 was also effective in inhibiting liver injury when administered as long as 3 h after or 1 h before alpha-Fas administration. Lastly, Western blot analysis demonstrated that processing of caspases 3, 6, and 8, as well as Bid (a protein responsible for the release of mitochondrial cytochrome C and amplification of the apoptotic cascade) was inhibited by IDN-1965. In conclusion, the broad-spectrum caspase inhibitor IDN-1965 is markedly effective at inhibiting Fas-mediated apoptosis by multiple routes of administration. The therapeutic potential of caspase inhibitors appears promising for the treatment of apoptosis-mediated liver injury based on potency and postinsult efficacy.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Chemical and Drug Induced Liver Injury/pathology , Cysteine Proteinase Inhibitors/pharmacology , Indoles/pharmacology , Oligopeptides/pharmacology , Alanine Transaminase/metabolism , Animals , Blotting, Western , Immunohistochemistry , Liver/pathology , Male , Mice , Mice, Inbred BALB C , fas Receptor/genetics
7.
Neuroscience ; 87(4): 905-11, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9759978

ABSTRACT

Modafinil is a novel wake-promoting compound for which the mechanism and sites of action are unknown. We examined the neural substrates in the brain for the actions of modafinil using 2-deoxyglucose autoradiography and compared the findings to those obtained with amphetamine. Modafinil showed a relatively restricted pattern of changes in brain regional metabolic activity, while amphetamine altered glucose utilization in a wide variety of brain regions. Both modafinil and amphetamine increased glucose utilization in all subregions of the hippocampus (subiculum, CA1-CA3 and dentate gyrus) and in the centrolateral nucleus of the thalamus. Modafinil also increased glucose utilization in the central nucleus of the amygdala, but amphetamine had no effect in this region. Brain structures in which amphetamine increased metabolic rate but modafinil had no effect included regions of the basal ganglia, other nuclei of the thalamus, the frontal cortex, the nucleus accumbens, the ventral tegmental area and the pontine reticular fields. These findings suggest that, while both modafinil and amphetamine promote wakefulness, they act via distinctly different mechanisms. Modafinil appears to act on a specific subset of brain pathways which regulate sleep and wakefulness, whereas amphetamine affects a greater number of cerebral structures involved in the regulation of these behavioral states. Modafinil also lacks the pronounced effects on the extrapyramidal motor system which are characteristic of amphetamine and other psychomotor stimulants, implying that the effects of modafinil are not mediated by the dopamine system and that modafinil may selectively increase wakefulness with fewer side effects.


Subject(s)
Amygdala/drug effects , Benzhydryl Compounds/pharmacology , Central Nervous System Stimulants/pharmacology , Deoxyglucose , Dextroamphetamine/pharmacology , Hippocampus/drug effects , Wakefulness/drug effects , Amygdala/metabolism , Animals , Brain/drug effects , Brain/metabolism , Energy Metabolism , Glucose/metabolism , Hippocampus/metabolism , Male , Modafinil , Motor Activity/drug effects , Motor Activity/physiology , Rats , Rats, Sprague-Dawley , Sleep/physiology
8.
J Comp Neurol ; 398(3): 373-92, 1998 Aug 31.
Article in English | MEDLINE | ID: mdl-9714150

ABSTRACT

Cell survival, death, and stress signals are transduced from the cell surface to the cytoplasm and nucleus via a cascade of phosphorylation events involving the mitogen-activated protein kinase (MAPK) family. We compared the distribution of p42 mitogen-activated protein kinase (p42MAPK) and its activator MAPK or ERK kinase (MEK1; involved in transduction of growth and differentiation signals), with c-Jun N-terminal kinase (JNK1) and its activator MEK4 (involved in transduction of stress and death signals) in the adult rat central nervous system. All four kinases were present in the cytoplasm, dendrites, and axons of neurons. The presence of p42MAPK and JNK1 in dendrites and axons, as well as in cell bodies, suggests a role for these kinases in phosphorylation and regulation of cytoplasmic targets. A high degree of correspondence was found between the regional distribution of MEK1 and p42MAPK. Immunostaining for MEK1 and p42MAPK was intense in olfactory structures, neocortex, hippocampus, striatum, midline, and interlaminar thalamic nuclei, hypothalamus, brainstem, Purkinje cells, and spinal cord. In addition to neurons, p42MAPK was also present in oligodendrocytes. Whereas MEK4 was ubiquitously distributed, JNK1 was more selective. Immunostaining for MEK4 and JNK1 was intense in the olfactory bulb, lower cortical layers, the cholinergic basal forebrain, most nuclei of the thalamus, medial habenula, and cranial motor nuclei. The distribution of MEK1 and p42MAPK proteins only partially overlapped with that of MEK4 and JNK1. This suggests that the growth/differentiation and death/stress pathways affected by these kinases may not necessarily act to counterbalance each other in response to extracellular stimuli. The differential distribution of these kinases may control the specificity of neuronal function to extracellular signals.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Central Nervous System/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Blotting, Western , Central Nervous System/enzymology , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Signal Transduction/physiology
9.
Neurosci Lett ; 241(2-3): 95-8, 1998 Jan 30.
Article in English | MEDLINE | ID: mdl-9507929

ABSTRACT

We examined the neuronal targets in the rat brain for the novel wakefulness-promoting agent modafinil and for amphetamine using c-Fos immunohistochemistry. Both modafinil and amphetamine induced neuronal expression of c-Fos-like immunoreactivity in the paraventricular nucleus of the hypothalamus, anterior hypothalamus and central nucleus of the amygdala. Modafinil also increased c-Fos-like immunoreactivity in the suprachiasmatic nucleus, while amphetamine had no effect. Brain regions in which amphetamine increased c-Fos-like immunoreactivity, but modafinil had no effect, included frontal cortex, striatum, lateral habenula, supraoptic nucleus and basolateral nucleus of the amygdala. These findings suggest that the mechanism of action of modafinil is different from that of amphetamine and that the neuronal targets for modafinil in the brain include nuclei of the hypothalamus and amygdala.


Subject(s)
Amphetamine/pharmacology , Benzhydryl Compounds/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Brain/cytology , Brain/metabolism , Immunohistochemistry , Male , Modafinil , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Wakefulness
10.
J Neurochem ; 70(2): 558-71, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453550

ABSTRACT

In vitro studies indicate that p42/p44MAPK phosphorylate both nuclear and cytoplasmic proteins. However, the functional targets of p42/p44MAPK activation in vivo remain unclear. To address this question, we localized activated p42/p44MAPK in hippocampus and cortex and determined their signaling effects after electroconvulsive shock treatment (ECT) in rats. Phosphorylated p42/p44MAPK content increased in the cytoplasm of hippocampal neurons in response to ECT. Consistent with this cytoplasmic localization, inhibition of ECT-induced p42/p44MAPK activation by the extracellular signal-regulated kinase kinase inhibitor PD098059 blocked phosphorylation of the cytoplasmic protein microtubule-associated protein 2c (MAP2c), but failed to inhibit the induction of the nuclear protein c-Fos in response to ECT. In contrast to hippocampal neurons, cortical neurons exhibited an increase in amount of phosphorylated p42/p44MAPK in both the nucleus and cytoplasm after ECT. Accordingly, PD098059 blocked the induction of Fos-like immunoreactivity in the nuclei of cortical neurons as well as MAP2c phosphorylation in the cytoplasm. Our data indicate that both nuclear and cytoplasmic substrates can be activated by p42/p44MAPK in vivo. However, the functional targets of p42/p44MAPK signaling depend on the precise location of p42/p44MAPK within different subcellular compartments of brain regions. These results indicate unique functional pathways of p42/p44MAPK-mediated signal transduction within different brain regions in vivo.


Subject(s)
Brain/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Signal Transduction/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Nucleus/physiology , Cerebral Cortex/physiology , Cytoplasm/metabolism , Electroshock , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hippocampus/physiology , MAP Kinase Kinase 1 , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase 3 , Organ Specificity , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley
11.
Exp Neurol ; 147(2): 418-27, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9344566

ABSTRACT

We evaluated the efficacy of insulin-like growth factor-1 (IGF-1) in attenuating neurobehavioral deficits following lateral fluid percussion (FP) brain injury. Male Sprague-Dawley rats (345-425 g, n = 88) were anesthetized and subjected to FP brain injury of moderate severity (2.4-2.9 atm). In Study 1, IGF-1 (1.0 mg/kg, n = 9) or vehicle (n = 14) was administered by subcutaneous injection at 15 min postinjury and similarly at 12-h intervals for 14 days. In animals evaluated daily for 14 days, IGF-1 treatment attenuated motor dysfunction over the 2-week period (P < 0.02). In Study 2, IGF-1 (4 mg/kg/day, n = 8 uninjured, n = 13 injured) or vehicle (n = 8 uninjured, n = 13 injured) was administered for 2 weeks via a subcutaneous pump implanted 15 min postinjury. IGF-1 administration was associated with increased body weight and mild, transient hypoglycemia which was more pronounced in brain-injured animals. At 2 weeks postinjury (P < 0.05), but not at 48 h or 1 week, brain-injured animals receiving IGF-1 showed improved neuromotor function compared with those receiving vehicle. IGF-1 administration also enhanced learning ability (P < 0.03) and memory retention (P < 0.01) in brain-injured animals at 2 weeks postinjury. Taken together, these data suggest that chronic, posttraumatic administration of the trophic factor IGF-1 may be efficacious in ameliorating neurobehavioral dysfunction associated with traumatic brain injury.


Subject(s)
Brain Injuries/drug therapy , Cognition Disorders/prevention & control , Insulin-Like Growth Factor I/therapeutic use , Movement Disorders/prevention & control , Neuroprotective Agents/therapeutic use , Animals , Brain Damage, Chronic/etiology , Brain Damage, Chronic/prevention & control , Brain Injuries/complications , Cognition Disorders/etiology , Infusion Pumps, Implantable , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Learning Disabilities/etiology , Learning Disabilities/prevention & control , Male , Maze Learning/drug effects , Movement Disorders/etiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Time Factors , Wounds, Nonpenetrating/drug therapy
12.
J Pharmacol Exp Ther ; 281(1): 522-30, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9103540

ABSTRACT

Recent studies indicate that a daily s.c. injection of 1 mg/kg of recombinant human insulin-like growth factor-1 (rhIGF-1) for 17 days is efficacious in enhancing the functional recovery of injured sciatic nerves in CD-1 mice. To identify and characterize surrogate marker(s) that are altered in association with the administration of an efficacious dose of rhIGF-1, dose-response curves (0.1, 1 and 10 mg/kg) and time course effects (0, 0.5, 3, 6 and 24 hr) were determined after acute (single) and chronic (once daily for 17 days) injections of rhIGF-1 in CD-1 mice. Plasma glucose levels decreased in a dose-dependent fashion after either acute or chronic injections of rhIGF-1 with maximal effects at 0.5 to 1 hr after administration of rhIGF-1. Among the three insulin-like growth factor binding proteins (IGFBPs) evaluated in the study, only IGFBP2 levels were consistently increased in a dose-dependent fashion with maximal effects 3 hr after the last of a series of injections of rhIGF-1. Furthermore, IGFBP2 levels increased at a dose of rhIGF-1 (1 mg/kg) that enhances the regeneration of injured sciatic nerves in mice. Chronic administration of insulin at doses that cause comparable decreases in plasma glucose to that of rhIGF-1 did not alter IGFBP2 levels or enhance hindlimb function suggesting that the beneficial effects of rhIGF-1 occur via activation of the type-I IGF receptor rather than the insulin receptor. Based on these criteria, IGFBP2 appears to be useful as a surrogate marker for determining the in vivo effects of rhIGF-1.


Subject(s)
Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor I/pharmacology , Animals , Biomarkers , Blood Glucose/analysis , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Male , Mice , Recombinant Proteins/pharmacology
13.
Biomed Pharmacother ; 51(4): 181-4, 1997.
Article in English | MEDLINE | ID: mdl-9207987

ABSTRACT

Aclacinomycin (ACM) is an oncostatic of the anthracycline family, largely used in patients and experimentally in mice. ACM has been reported to enhance phagocytosis, secretion of free oxygen radicals and of interleukin 1. Its injection is also followed by an increase of the cytotoxic and cytostatic activity of murine peritoneal macrophages. In the present work we investigated whether ACM modifies the antigen-presenting cell capacity of murine peritoneal macrophages. Purified T lymphocytes were cultured with peritoneal macrophages from either normal or ACM treated mice (4 mg/kg day -4) which were previously incubated with phytohemagglutinin. The T cell proliferative response was greater in cultures with normal macrophages, indicating that macrophages from ACM-treated mice had a better antigen presenting activity than normal untreated macrophages.


Subject(s)
Aclarubicin/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antigen-Presenting Cells/immunology , Macrophages, Peritoneal/immunology , Animals , Immunity, Cellular/drug effects , Male , Mice , Mice, Inbred BALB C
14.
Brain Res ; 774(1-2): 20-6, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9452187

ABSTRACT

Vincristine is a commonly used antitumor agent whose major dose-limiting side-effect is a mixed sensorimotor neuropathy. To assess whether insulin-like growth factor-I (IGF-I), a neurotrophic agent that supports the survival of motoneurons and enhances regeneration of motor and sensory neurons, could prevent the peripheral neuropathy produced by vincristine, mice were treated with both vincristine (1.7 mg/kg, i.p., 2 x /week) and/or IGF-I (0.3 or 1 mg/kg, s.c. daily) for 10 weeks. In mice treated with vincristine alone, there was evidence of a mixed sensorimotor neuropathy as indicated by changes in behavior, nerve conduction and histology. Caudal nerve conduction velocity was significantly slower in mice treated with vincristine alone as compared with vehicle-treated mice. Vincristine treatment alone also significantly increased hot-plate latencies and reduced gait support and stride length, but not toe spread distances. The effects of vincristine were accompanied by degeneration of sciatic nerve fibers and demyelination, indicating a peripheral neuropathy. IGF-I (1 mg/kg, s.c.) administered to vincristine-treated mice prevented the neurotoxic effects of vincristine as measured by nerve conduction, gait, response to noxious stimuli and nerve histology. At a lower dose of 0.3 mg/kg administered s.c., IGF-I partially ameliorated the neuropathy induced by vincristine as this dose only prevented the change in nerve conduction and hot-plate latencies. IGF-I administered alone had no effect on any of these parameters. These results suggest that IGF-I prevents both motor and sensory components of vincristine neuropathy and may be useful clinically in preventing the neuropathy induced by vincristine treatment.


Subject(s)
Antineoplastic Agents, Phytogenic , Insulin-Like Growth Factor I/pharmacology , Nervous System Diseases/chemically induced , Nervous System Diseases/prevention & control , Vincristine , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Gait/drug effects , Male , Mice , Mice, Inbred Strains , Movement/drug effects , Movement/physiology , Nervous System Diseases/physiopathology , Neural Conduction/drug effects , Pain/physiopathology , Reaction Time/drug effects , Sensation/drug effects , Sensation/physiology
15.
J Neurosci ; 16(13): 4146-54, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8753876

ABSTRACT

Although the interleukin-1beta converting enzyme (ICE)/CED-3 family of proteases has been implicated recently in neuronal cell death in vitro and in ovo, the role of specific genes belonging to this family in cell death in the nervous system remains unknown. To address this question, we examined the in vivo expression of one of these genes, Ice, after global forebrain ischemia in gerbils. Using RT-PCR and Western immunoblot techniques, we detected an increase in the mRNA and protein expression of ICE in hippocampus during a period of 4 d after ischemia. Chromatin condensation was observed in CA1 neurons within 2 d after ischemia. Internucleosomal DNA fragmentation and apoptotic bodies were observed between 3 and 4 d after ischemia, a period during which CA1 neuronal death is maximal. In nonischemic brains, ICE-like immunoreactivity was relatively low in CA1 pyramidal neurons but high in scattered hippocampal interneurons. After ischemia, ICE-like immunoreactivity was not altered in these neurons. ICE-like immunoreactivity, however, was observed in microglial cells in the regions adjacent to the CA1 layer as early as 2 d after ischemic insult. The increase in ICE-like immunoreactivity was robust at 4 d after ischemia, a period that correlates with the DNA fragmentation observed in hippocampal homogenates of ischemic brains. These results provide the first evidence for the localization and induction of ICE expression in vivo after ischemia and suggest an indirect role for ICE in ischemic damage through mediation of an inflammatory response.


Subject(s)
Brain Ischemia/enzymology , Cysteine Endopeptidases/metabolism , Hippocampus/enzymology , Animals , Apoptosis , Base Sequence , Caspase 1 , Cysteine Endopeptidases/genetics , Gerbillinae , Hippocampus/pathology , Immunohistochemistry , Male , Microglia/enzymology , Molecular Probes/genetics , Molecular Sequence Data , Neurons/pathology , RNA, Messenger/metabolism , Time Factors , Tissue Distribution
16.
Ciba Found Symp ; 196: 18-27; discussion 27-38, 1996.
Article in English | MEDLINE | ID: mdl-8866126

ABSTRACT

Neuromuscular/neurodegenerative disorders, such as the death of spinal cord motor neurons in amyotrophic lateral sclerosis (ALS) or the degeneration of spinal cord motor neuron axons in certain peripheral neuropathies, present a unique opportunity for therapeutic intervention with neurotrophic proteins. We have found that in mixed rat embryonic spinal cord cultures or in purified motor neuron preparations, recombinant human insulin-like growth factor 1 (rhIGF-1) enhances the survival of motor neurons at EC50 concentrations of 2 nM, consistent with an interaction at the tyrosine kinase-coupled rhIGF-1 receptor. In a model of programmed cell death in ovo, administration of rhIGF-1 produces a marked survival of motor neurons. In a variety of models of predominantly motor neuron or nerve injury in rodents, administration of rhIGF-1 prevents the death of motor neurons in neonatal facial nerve lesions, attenuates the loss of cholinergic phenotype in adult hypoglossal nerve axotomy and hastens recovery from sciatic nerve crush in mice. In a genetic model of motor neuron compromise, the wobbler mouse, rhIGF-1 (1 mg/kg s.c. daily) delayed the deterioration of grip strength and provided for a more normal distribution of fibre types. In addition, rhIGF-1 (0.3-1.0 mg/kg s.c. daily) prevents the motor and/or sensory neuropathy in rodents caused by vincristine, cisplatinum or Taxol. These combined data indicate that rhIGF-1 has marked effects on the survival of compromised motor neurons and the maintenance of their axons and functional connections. They also suggest the potential utility of rhIGF-1 for the treatment of diseases such as ALS and certain neuropathies.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Insulin-Like Growth Factor I/pharmacology , Motor Neurons/drug effects , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Female , Humans , Insulin-Like Growth Factor I/metabolism , Mice , Motor Neurons/cytology , Motor Neurons/metabolism , Nerve Degeneration , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Spinal Cord/embryology
17.
J Pharmacol Exp Ther ; 274(3): 1443-9, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7562520

ABSTRACT

Despite numerous reports suggesting that insulin-like growth factor-I (IGF-I) may be involved in the survival and regeneration of damaged neurons in vitro and after local administration in vivo, there have been few studies on the effect of IGF-I administered systemically on regeneration of damaged nerves in vivo and the functional consequences of enhanced regeneration. In an earlier study, recombinant human IGF-I (rhIGF-I) administered systemically enhanced the rate of regeneration after a sciatic crush as measured by the number of nerve fibers/muscle section. The purpose of this study was to follow up this finding by evaluating whether rhIGF-I administered peripherally enhances the rate of functional recovery. In this study following nerve injury, mice lost the ability to grip a wire screen with their hind paws and to walk normally as indicated by a decrease in toe spread, internal toe spread and an increase in the angle between hind feet. The ability of injured mice treated with rhIGF-I to grip an inverted screen returned to control levels significantly faster than that of vehicle-treated mice (day 12 vs. day 15, respectively). Similarly, rhIGF-I treatment of injured mice resulted in toe spread, internal toe spread and angle values that were significantly better than that of vehicle-treated mice and returned to control levels faster than vehicle-treated injured mice. There was a parallel loss of innervation after sciatic nerve crush as measured by a loss in choline acetyltransferase activity in the soleus and gastrocnemius muscles.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Nerve Regeneration , Sciatic Nerve/physiology , Animals , Dose-Response Relationship, Drug , Humans , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacokinetics , Male , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Sciatic Nerve/injuries
18.
Exp Neurol ; 124(1): 73-88, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8282084

ABSTRACT

Motor neuronal disorders, such as the loss of spinal cord motor neurons in amyotrophic lateral sclerosis or the degeneration of spinal cord motor neuron axons in certain peripheral neuropathies, present a unique opportunity for therapeutic intervention with neurotrophic proteins. Normally, such proteins do not cross the blood-brain barrier, but spinal cord motor neuron axons and nerve terminals lie outside the barrier and thus may be targeted by systemic administration of protein growth factors. Insulin-like growth factor-I (IGF-I) receptors are present in the spinal cord, and, like members of the neurotrophin receptor family, IGF-I receptors mediate signal transduction via a tyrosine kinase domain. IGF-I was found to prevent the loss of choline acetyltransferase activity in embryonic spinal cord cultures, as well as to reduce the programmed cell death of motor neurons in vivo during normal development or following axotomy or spinal transection. Consistent with earlier reports that IGF-I enhances motor neuronal sprouting in vivo, subcutaneous administration of IGF-I increases muscle endplate size in rats. Subcutaneous injections of IGF-I also accelerate functional recovery following sciatic nerve crush in mice, as well as attenuate the peripheral motor neuropathy induced by chronic administration of the cancer chemotherapeutic agent vincristine in mice. Doses of IGF-I that accelerate recovery from sciatic nerve crush in mice result in elevated serum levels of IGF-I which are similar to those obtained following subcutaneous injections of formulated recombinant human IGF-I (Myotrophin) in normal human subjects. Based on these findings, together with evidence of safety in animals and man, clinical trials of recombinant human IGF-I have been initiated in patients with amyotrophic lateral sclerosis and are planned to begin soon in patients with chemotherapy-induced peripheral neuropathies.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Insulin-Like Growth Factor I/therapeutic use , Motor Neuron Disease/drug therapy , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Survival , Humans , Insulin-Like Growth Factor I/pharmacology , Models, Biological , Molecular Sequence Data , Motor Neuron Disease/physiopathology , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/physiology , Nervous System/drug effects , Nervous System Physiological Phenomena , Receptor, IGF Type 1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL