Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Osteoarthritis Cartilage ; 25(1): 46-52, 2017 01.
Article in English | MEDLINE | ID: mdl-27592041

ABSTRACT

OBJECTIVE: This publication summarizes the clinical development of the compound SAR113945, an IκB kinase inhibitor injected intra-articularly in a slow-release formulation to treat patients with symptomatic osteoarthritis (OA) of the knee. RESULTS: In vitro experiments demonstrated a specific inhibition of the IκB kinase complex. Profiling of SAR113945 on kinases, enzymes and ion channels supported the initiation of a clinical development. Cellular assay systems also revealed an inhibition in the synthesis of interleukin 1ß, tumor necrosis factor α (TNFα) and the prostaglandin E2 (PGE2). In vivo studies demonstrated positive effects of SAR113945 on thermal and mechanical hyperalgesia and even showed superiority in comparison with triamcinolone. Pharmacokinetic measurements showed a sustained release of dissolved SAR113945 locally supporting a comparably high exposure in the knee joint combined with a low systemic exposure. Three phase 1 studies with a dose-escalating design confirmed safety and tolerability of SAR113945. In those studies SAR113945 showed a positive trend on the WOMAC scores. The proof-of-concept or phase 2a study failed to show any effect in the overall group of recruited study participants for the primary endpoint, the WOMAC pain subscore at day 56, but showed a statistically significant difference in a subgroup of patients who had presented with effusion at baseline. CONCLUSION: Inhibiting the NFκB signaling pathway is an attractive method to treat patients with signs and symptoms of OA. The preclinical work and the results of the phase 1 studies appeared promising for a full clinical development, however, the proof-of-concept study failed to show efficacy in a larger patient sample size.


Subject(s)
I-kappa B Kinase/antagonists & inhibitors , Osteoarthritis, Knee/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Delayed-Action Preparations , Humans , Injections, Intra-Articular , Knee Joint , Male , Proof of Concept Study , Rats , Rats, Long-Evans , Triamcinolone/therapeutic use
3.
Osteoarthritis Cartilage ; 11(10): 738-46, 2003 Oct.
Article in English | MEDLINE | ID: mdl-13129693

ABSTRACT

OBJECTIVE: To study the effect of pralnacasan, the orally bioavailable pro-drug of a potent, non-peptide inhibitor of interleukin-1beta converting enzyme (ICE), RU 36384/VRT-18858, on joint damage in two mouse models of knee osteoarthritis (OA). DESIGN: In a collagenase-induced OA model, pralnacasan was given orally by gavage to female Balb/c mice at 0, 12.5, 25 and 50 mg/kg twice a day. In the second study, pralnacasan was tested in male STR/1N mice, which develop OA spontaneously, by administering food-drug mixtures ad libitum at concentrations of 0, 700 and 4200 ppm (mg/kg food). OA joint damage was assessed by a semi-quantitative histopathological score in both studies. In the STR/1N mouse study, urinary levels of collagen cross-links hydroxylysylpyridinoline (HP) and lysylpyridinoline (LP) were determined by high-pressure liquid chromatography at baseline, after 3 and 6 weeks of treatment and RU 36384/VRT-18858 plasma concentrations was measured after 6 weeks. RESULTS: In both studies, the mice developed moderate to severe knee joint OA in the medial joint compartments (tibial plateau and femoral condyle), the non-treated control groups showing median histopathological scores from 18 to 21 of a maximal score of 32. Pralnacasan was well tolerated. At the doses of 12.5 and 50 mg/kg in collagenase-induced OA and at the high dose of 4200 ppm in STR/1N mice pralnacasan treatment significantly reduced OA by 13-22%. In the STR/1N mice, urinary levels of HP cross-links and the ratio of HP/LP, which are indicators of joint damage in OA, were significantly reduced in the high dose group by 59 and 84%, respectively. CONCLUSIONS: The ICE inhibitor pralnacasan reduced joint damage in two experimental models of OA and has the potential to become a disease-modifying drug for the treatment of OA.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Azepines/therapeutic use , Caspase Inhibitors , Isoquinolines/therapeutic use , Osteoarthritis, Knee/drug therapy , Pyridazines/therapeutic use , Amino Acids/metabolism , Animals , Body Weight , Collagen/analysis , Disease Models, Animal , Eating , Female , Hindlimb , Joints/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology
4.
Eur J Pharmacol ; 382(1): 27-33, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10556501

ABSTRACT

Increased beta-amyloid production is believed to play a central role in the pathogenesis of Alzheimer's disease. Amyloid is deposited not only in the brain of Alzheimer patients as senile plaques but also in the cerebral vessel wall leading to cerebral amyloid angiopathy. Freshly solubilised amyloid beta-(1-40) was previously reported to exert a vasoconstrictor effect. We investigated whether amyloid beta-(1-40) affects the nitric oxide (NO)/cyclic GMP pathway in primary cultured endothelial cells from bovine aorta and rat coronary microvessels. Surprisingly, a significant increase in cyclic GMP production after incubation with freshly dissolved amyloid beta-(1-40) was found. The stimulation of cyclic GMP production could be inhibited by the bradykinin B(2) receptor antagonist icatibant, the NO synthase inhibitor N-omega-nitro-L-arginine, the serine protease inhibitor 3, 4-dichloroisocoumarin and the selective plasma kallikrein inhibitor Pefabloc PK, suggesting activation of the plasma kallikrein-kinin system. This is supported by a three- to four-fold increase in kinins in the supernatant of both types of endothelial cells after incubation with amyloid beta-(1-40) at concentrations of 10(-7) and 10(-6) mol/l.


Subject(s)
Amyloid beta-Peptides/pharmacology , Cyclic GMP/biosynthesis , Endothelium, Vascular/drug effects , Kinins/metabolism , Peptide Fragments/pharmacology , Amyloid beta-Peptides/physiology , Animals , Aorta/cytology , Aorta/drug effects , Aorta/ultrastructure , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Cattle , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/ultrastructure , Coumarins/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/ultrastructure , Enzyme Inhibitors/pharmacology , Isocoumarins , Kallikreins/antagonists & inhibitors , Microcirculation/drug effects , Microscopy, Electron , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Peptide Fragments/physiology , Rats , Serine Proteinase Inhibitors/pharmacology
5.
Neurosci Lett ; 260(1): 17-20, 1999 Jan 22.
Article in English | MEDLINE | ID: mdl-10027689

ABSTRACT

This study examines the effect of Propentofylline (PPF) on reactive microglia in the lumbar spinal cord in rats following focal cerebral ischaemia produced by permanent occlusion of the middle cerebral artery (MCA). Our results showed that daily treatment of PPF beginning at 24 h after MCA occlusion for 2 or 4 consecutive days markedly suppressed the microglial response as detected immunohistochemically with OX-42. The most dramatic effect was the prevention of transformation of ramified microglia into amoeboidic form as well as formation of perineuronal microglia in close association with the soma of motoneurons. This has greatly amplified the potentiality of PPF used as a neuroprotective drug against microglia-related neuron damage induced by cerebral ischaemia.


Subject(s)
Microglia/drug effects , Neuroprotective Agents/pharmacology , Spinal Cord/drug effects , Xanthines/pharmacology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cerebral Arterial Diseases/drug therapy , Cerebral Arterial Diseases/metabolism , Cerebral Arterial Diseases/pathology , Male , Microglia/physiology , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar , Spinal Cord/metabolism , Spinal Cord/physiology , Xanthines/therapeutic use
6.
J Neural Transm Suppl ; 54: 167-74, 1998.
Article in English | MEDLINE | ID: mdl-9850925

ABSTRACT

A significant role of a pathological glial cell activation in the pathogenesis of Alzheimer's disease is supported by the growing evidence that inflammatory proteins, which are produced by reactive astrocytes, promote the transformation of diffuse beta-amyloid deposits into the filamentous, neurotoxic form. A number of vicious circles, driven by the release of TNF-a and free oxygen radicals from microglial cells, may cause an upregulated microglial activation and their production of interleukin-1 which triggers, secondarily, the crucial activation of astrocytes. Reactive functional changes of glial cells seem to be controlled by an altered balance of the second messengers Ca2+ and cAMP and can be counterregulated by the endogenous cell modulator adenosine which strengthens the cAMP-dependent signalling chain. A further reinforcement of the homeostatic adenosine effects on glial cells by pharmaca, such as propentofylline, may add to neuroprotection in Alzheimer's disease.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Brain/immunology , Brain/pathology , Neuroglia/pathology , Adenosine/physiology , Alzheimer Disease/physiopathology , Astrocytes/pathology , Astrocytes/physiology , Brain/physiopathology , Calcium/metabolism , Cyclic AMP/metabolism , Humans , Interleukin-1/physiology , Microglia/pathology , Microglia/physiology , Models, Neurological , Neuroglia/immunology , Second Messenger Systems , Tumor Necrosis Factor-alpha/physiology
7.
Alzheimer Dis Assoc Disord ; 12 Suppl 2: S21-8, 1998.
Article in English | MEDLINE | ID: mdl-9769026

ABSTRACT

Cascading glial cell activation is believed to play an essential pathogenic role in the development of dementia. Reactive microglia may contribute to neuronal damage by the generation of free oxygen radicals and nitric oxide (NO), which forms the particularly aggressive peroxynitrites, and by the release of potentially neurotoxic cytokines such as tumor necrosis factor-alpha (TNF-alpha). The pathologically stimulated release of interleukin-1beta (IL-1beta) from microglial cells triggers secondary activation of astrocytes, which are forced to proliferate and to give up their differentiated state. As a consequence, physiologically required astrocyte functions may be impaired, such as uptake of glutamate and K+ from the extracellular space and release of neurotrophic factors. At the same time, production of inflammatory proteins which, for example, promote the formation of toxic beta-amyloids, is reported to be stimulated in reactive astrocytes. Because the complex molecular signaling that controls glial cell activation is only beginning to be elaborated, we attempted to elucidate the role that has been adopted during evolution by the endogenous cell modulator adenosine. This nucleoside exerts a homeostatic effect on reactive glial cell functions by a sophisticated control of the second messenger interplay, counteracting a pathologically induced dysbalance of the Ca2+- and cAMP-dependent signaling. A strengthening of the cAMP-dependent signaling chains was found to counteract the proliferation rate, the formation of free oxygen radicals, and the stimulated release of TNF-alpha and IL-1beta in cultivated microglia. It also helped proliferative astrocytes to regain their differentiated state and a mature ion channel pattern. The cAMP-linked homeostatic adenosine effects could be reinforced or mimicked by propentofylline, a pharmacon that raises the effective extracellular concentration of adenosine by inhibiting its cellular reuptake and increases the cellular cyclic nucleotide content by selective phosphodiesterase inhibition. We conclude that a pharmacologically reinforced homeostatic control of the pathologically altered Ca2+/cAMP crosstalk may prevent glia-related neuronal damage, providing a potential option for the treatment of dementia.


Subject(s)
Alzheimer Disease/immunology , Astrocytes/immunology , Macrophage Activation/immunology , Microglia/immunology , Neuroprotective Agents/pharmacology , Xanthines/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Astrocytes/drug effects , Calcium/metabolism , Cytokines/physiology , Homeostasis/drug effects , Humans , Macrophage Activation/drug effects , Microglia/drug effects , Second Messenger Systems/drug effects , Second Messenger Systems/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
8.
Alzheimer Dis Assoc Disord ; 12 Suppl 2: S15-20, 1998.
Article in English | MEDLINE | ID: mdl-9769025

ABSTRACT

Ameboid microglia are activated macrophages in the developing brain. With age, these cells undergo gradual transformation into the adult form, known as ramified or resting microglia. In response to neuronal insults, microglia change their morphology and immunophenotype and proliferate to become full-blown brain macrophages. Microglia release a battery of neurotoxic substances. Responses to neuronal damage occur at various intervals after the insult, suggesting that microglia may be an attractive target for pharmacologic intervention. The cerebrospinal fluid (CSF) of Alzheimer disease (AD) patients contains antibodies that recognize activated microglia in the developing rat and in the ischemic gerbil brain. These results suggest that AD shares common mechanisms related to the activation of microglia with both these experimental models. In vitro, the xanthine derivative propentofylline (PPF) depresses the production of reactive oxygen intermediates produced by macrophages. To appreciate in vivo interactions of PPF, two models were employed: developing rats and adult gerbils exposed to ischemia. Newborn rats were administered PPF (10 mg/kg) for 7 days. Gerbils were exposed to 5 min of transient forebrain ischemia and received PPF (10 mg/kg) 24 h later until the day before sacrifice. Animals were sacrificed at 7 or 14 days after reperfusion. Brains were processed for immunocytochemistry. Reactive microglia were visualized with monoclonal antibodies OX18 and OX42 or AD-CSF microglia antibodies. In the case of ischemia, an antibody against the amyloid precursor protein (APP) (residues 676-695) was included. Newborn rats receiving PPF for 7 days displayed a dramatic reduction in the number of activated microglia compared with untreated littermates. Ischemic control in gerbils showed complete nerve death, accumulations of APP, and enhanced microglial reactivity. In gerbils receiving PPF, APP accumulation was absent or very slight, and activated microglia were downregulated. The ability of PPF to interfere with activated microglia suggests that this agent may be useful for slowing progressive nerve cell death associated with AD, which is considered to be largely influenced by pathologic glial reactions.


Subject(s)
Alzheimer Disease/immunology , Macrophage Activation/immunology , Microglia/immunology , Neuroprotective Agents/pharmacology , Xanthines/pharmacology , Adult , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Brain Ischemia/immunology , Cholinergic Fibers/drug effects , Cholinergic Fibers/immunology , Disease Models, Animal , Gerbillinae , Humans , Macrophage Activation/drug effects , Microglia/drug effects , Prosencephalon/blood supply , Prosencephalon/immunology , Rats
9.
Article in German | MEDLINE | ID: mdl-9931990

ABSTRACT

Propentofylline (Karsivan, Hoechst Roussel Vet) is a selective inhibitor of adenosine transport and phosphodiesterase. For several years it has been well established in the geriatric therapy of the dog improving hemodynamics in cerebral and peripheral compartments. In human medicine clinical development of this pharmaceutical has already entered an advanced stage for the long-term therapy of patients with Alzheimer's disease and vascular dementia. In the brains of senile dogs and in human patients suffering from Alzheimer's disease comparable neuropathological findings can be made. In senile dogs a distinctive correlation exists between the quantity of beta-amyloid accumulation and the degree of dementia. The extension of knowledge by clinical studies in humans and by experimental studies in animals may contribute to a deeper understanding of therapeutical approaches of cognitive dysfunction in the old dog. The xanthine derivative propentofylline [1-(5'-oxohexyl)-3-methyl-7-propylxanthine] directly interfers with the neurodegenerative process and reduces the extent of damage to brain structures. In experimental models of vascular dementia and/or Alzheimer's disease it improves cognitive functions, inhibits inflammatory processes as well as excessive activation of microglia, formation of free radicals, cytocines and abnormal amyloid precursor proteins (APP). It stimulates synthesis and liberation of nerve growth factor (NGF) and reduces ischemic damage to the brain. In clinical studies in humans it improved cognitive functions as well as global functions and the ability to cope with tasks of routine daily life in patients suffering from Alzheimer's disease and vascular dementia.


Subject(s)
Cerebrovascular Circulation/drug effects , Cognition Disorders/drug therapy , Dog Diseases/drug therapy , Hemodynamics/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Xanthines/pharmacology , Xanthines/therapeutic use , Adenosine/antagonists & inhibitors , Alzheimer Disease/drug therapy , Animals , Dementia, Vascular/drug therapy , Dog Diseases/psychology , Dogs , Humans , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use
10.
Ann N Y Acad Sci ; 825: 1-10, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9369970

ABSTRACT

As illustrated in Figure 1, a disturbance of the intracellular Ca2+ homeostasis is thought to be a common pathogenic factor for the generation of secondary nerve cell damage that develops after brain trauma or stroke or during the course of neurodegenerative diseases. A neuronal Ca2+ overload which may result from an excessive glutamate-evoked membrane depolarization and consecutive Ca2+ influx as well as from an activation of metabotropic receptors and consecutive intracellular Ca2+ mobilization is known to have direct toxic effects on the cytoskeleton and the cell metabolism of neurons. In addition, a Ca(2+)-dependent activation of glial cells along with the loss of physiologically required mature astrocyte functions and with the acquisition of potentially neurotoxic microglial properties, has more recently been recognized as an additive pathogenic factor. This may provide an effective target for pharmacological interference. Specifically, the reinforcement of an endogenous homeostatic regulator, which obtained its sophisticated know-how during evolution, may provide a neuroprotective therapy which can handle the complexity of the pathological process with a minor risk of pharmacological side effects. Adenosine is such an ancient molecular signal that acts on both neurons and glial cells. In neurons, adenosine activates K+ and Cl- conductances, which limits synaptically evoked depolarization, thus counteracting the Ca2+ influx through voltage-dependent and NMDA receptor-operated ion channels. This A1 receptor-mediated effect seems to be the major action by which adenosine adds directly to the protection of neurons against Ca(2+)-dependent damage. In glial cells, the prevalent effect of adenosine is its regulatory influence on the Ca2+ and cAMP-dependent molecular signaling that determines the cellular proliferation rate, the differentiation state and related functions. When mimicking the activation of metabotropic glutamate receptors in cultures of immature rat astrocytes, which largely resemble pathologically activated astrocytes, a transient Ca2+ mobilization was initiated by adenosine. This A1 receptor-mediated Ca2+ signal caused a prolonged potentiation of the A2 receptor-mediated intracellular cAMP rise. An experimentally sustained enhancement of the cAMP signaling initiated the differentiation of cultured astrocytes and the new expression of K+ and Cl- channels which are required for the physiological astrocyte function to maintain the extracellular ion homeostasis. Evidence is accumulating that a strengthening of the cAMP signaling, which can be achieved by adenosine agonists and also by the pharmacon propentofylline (an adenosine uptake blocker and phosphodiesterase inhibitor), stimulates the mRNA production of neurotrophic factors in astrocytes. In cultured microglial cells, several days' treatment with adenosine agonists or propentofylline markedly inhibited their proliferation rate, the in vitro spontaneously occurring transformation into macrophages and their particularly high formation of free oxygen radicals. Adenosine agonists also depressed the release of the potentially toxic cytokine TNF alpha and induced programmed cell death in immunologically activated microglial cells. We conclude that a pharmacological reinforcement of the endogenous cell modulator adenosine may provide neuroprotection by counteracting neuronal Ca2+ overload, by depressing potentially neurotoxic microglial functions and by regaining physiologically required properties of differentiated astrocytes. Further information about the influence of adenosine on the molecular signaling and on ischemic brain damage is given in Refs. 37 and 38, and about the implicated possible relevance for the treatment of stroke in Ref. 39.


Subject(s)
Adenosine/pharmacology , Brain/physiology , Neuroglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/physiology , Brain/cytology , Calcium/metabolism , Cyclic AMP/metabolism , Humans , Neuroglia/cytology , Neuroglia/physiology , Neurons/cytology , Neurons/physiology , Rats
11.
Ann N Y Acad Sci ; 826: 337-47, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9329704

ABSTRACT

A pathological glial cell activation, which forces microglia to transform into immunocompetent cells with cytotoxic properties and astrocytes to "de-differentiate," presumably adds to neurodegenerative diseases. We examined the modulatory effect of adenosine on the Ca2+ and cAMP-dependent regulation of such reactive glial cell properties in culture and tested possibilities of pharmacologic reinforcement. A strengthening of the cAMP-signaling, as could be achieved by adenosine agonists via a Ca(2+)-dependent action, favored the differentiation of proliferating astrocytes and associated neuroprotective properties (ion homeostasis, formation of trophic factors). But potentially neurotoxic properties of microglial cells were inhibited. Adenosine depressed their proliferation rate and transformation into macrophages, their particularly high formation of reactive oxygen intermediates and the release of the cytokine TNF-alpha. Similar effects were obtained with propentofylline, which acts as selective cAMP/cGMP phosphodiesterase inhibitor and also increases the effective concentration of adenosine by blocking its cellular reuptake. The recently observed induction of microglial apoptosis by elevated extracellular adenosine levels may further contribute to limit secondary nerve cell damage related to a pathological glial cell activation.


Subject(s)
Alzheimer Disease/drug therapy , Homeostasis , Neuroglia/physiology , Signal Transduction , Xanthines/therapeutic use , Adenosine/physiology , Astrocytes/physiology , Calcium/metabolism , Cyclic AMP/metabolism , Humans , Models, Biological , Molecular Mimicry , Second Messenger Systems
12.
Behav Brain Res ; 83(1-2): 123-8, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9062670

ABSTRACT

Dementia is one of the major medical challenges in the industrialized countries. An increasing number of demented patients shows ischemic brain lesions. One strategy to treat dementia could be the reinforcement of the multifarious neuroprotective actions of the endogenous cell modulator adenosine. This involves the modulation of neuronal and glial cell functions. Adenosine appears to raise the threshold for the initiation of the pathophysiological cascade of ischemic neuronal death by counteracting ischemic membrane depolarization and the successive disturbance of intracellular Ca2+ homeostasis. When this threshold is overcome, adenosine seems to stimulate astrocyte differentiation and reinforce important protective astrocyte functions, e.g., synthesis and release of neurotrophic factors such as NGF and the clearance of abnormal neurotoxic levels of K+ and glutamate from the extracellular space. Propentofylline, a combined inhibitor of adenosine reuptake and cAMP phosphodiesterases, reinforces the actions of endogenous adenosine and cAMP. One important action of propentofylline is the inhibition of potentially neurotoxic functions of activated microglia (free radical formation and transformation into brain macrophages). Such drugs may help to inhibit the progressive neurodegenerative process in dementia.


Subject(s)
Adenosine/therapeutic use , Dementia, Vascular/drug therapy , Neuroglia/drug effects , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Adenosine/pharmacology , Animals , Dementia, Vascular/pathology , Humans , Neuroprotective Agents/pharmacology , Purinergic P1 Receptor Antagonists
13.
Mol Chem Neuropathol ; 28(1-3): 185-90, 1996.
Article in English | MEDLINE | ID: mdl-8871958

ABSTRACT

In view of the increasing evidence that a pathological glial activation plays a significant role in the development of neurodegenerative diseases, we investigated the underlying molecular signaling as a possible target for the pharmacological therapy. Here, we are particularly focusing on the endogenous modulation of the CA2+ and cyclic nucleotide-dependent signaling by the nucleoside adenosine and its reinforcement by the xanthine derivative propentofylline (PPF). As an experimental model, we used cultured rat microglial cells and astrocytes that are immature, show a high proliferation rate, and resemble in several aspects pathologically activated glial cells. A prolonged increase of the cellular cAMP level favored the differentiation of cultured astrocytes and associated properties required for the physiological nerve cell function. On the other hand a strengthening of the cyclic nucleotide-dependent signaling inhibited potentially neurotoxic properties of cultured microglial cells. Similar effects were obtained by treatment with propentofylline, which mimicked modulatory adenosine effects and increased the intracellular level of cAMP and cGMP. Such a pharmacological glial cell conditioning, obtained by modifying the strength and the timing of these second messengers, may provide a therapy of neurodegenerative diseases in which a pathological activation of microglial cells and astrocytes is discussed to playa pathogenic role.


Subject(s)
Adenosine/physiology , Microglia/physiology , Neuroglia/physiology , Neuroprotective Agents , Signal Transduction , Xanthines/pharmacology , Animals , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Cyclic AMP/metabolism , Microglia/cytology , Microglia/drug effects , Neuroglia/drug effects , Rats , Signal Transduction/drug effects
14.
Exp Neurol ; 137(2): 345-9, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8635550

ABSTRACT

Propentofylline is a xanthine derivative that has been known to protec t neurons against ischemia-induced damage. To assess its neuroprotective mechanisms, we examined the effect of propentofylline on microglial proliferation that is thought to play an important role in neuronal damage. We determined the proliferation of microglia cultured from neonatal rat brains by measuring [3H]thymidine update. Propentofylline inhibited microglial proliferation in a dose dependent manner; EC50 was about 3 mu M. Similar results were observed with 2-chloroadenosine (agonist for A1 and A2 adenosine receptors) and 2-chloro-N6-cyclopentyladenosine (A1 receptor agonist) but not with 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethyl-carboxamidoadenosine hydrochloride (A2 receptor agonist). However, 8-cyclopentyl-1,3-dipropylxathine (A1 receptor antagonist) could not reverse the inhibitory effect of propentofylline. Our results suggest that the neuroprotection by propentofylline is, as least in part, due to the direct effect of the drug on microglia and that the drug inhibits the proliferation via a certain mechanism not directly mediated by adenosine receptors.


Subject(s)
Adenosine/pharmacology , Anti-Ulcer Agents/pharmacology , Microglia/drug effects , Xanthines/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Microglia/physiology , Radioligand Assay , Rats , Thymidine/metabolism
15.
Ann N Y Acad Sci ; 777: 404-9, 1996 Jan 17.
Article in English | MEDLINE | ID: mdl-8624121

ABSTRACT

The pharmacological profile of HWA 285 favors its use in patients with both Alzheimer's disease (PDD) and/or vascular dementia (MID). Clinical trials showed clinically relevant, statistically significant efficacy in the domains of cognitive function, global function and activities of daily living (ADL) in both PDD and MID. HWA 285 had a prolonged symptomatic effect for at least 12 months, although therapeutic effects were seen already after the first 3 months of treatment. HWA 285 was very well tolerated for at least 1 year.


Subject(s)
Alzheimer Disease/drug therapy , Dementia, Vascular/drug therapy , Xanthines/therapeutic use , Activities of Daily Living , Alzheimer Disease/physiopathology , Brain Ischemia/drug therapy , Clinical Trials as Topic , Dementia, Vascular/physiopathology , Humans , Treatment Outcome
16.
Int J Biochem ; 26(10-11): 1227-36, 1994.
Article in English | MEDLINE | ID: mdl-7531656

ABSTRACT

Adenosine is released during brain ischemia and provides neuroprotection by actions on nerve and glial cells. Activation of the adenosine A1 receptor enhances the K+ and Cl- conductance in neurons, leading to membrane hyperpolarization and postsynaptic reduction of neuronal Ca2+ influx through voltage- and NMDA receptor-dependent channels. In addition adenosine A1 receptor activation decreases excitatory amino acid release, possibly via inhibition of N- and P-type Ca2+ channels. The A1 and A2 receptors, coupled to Gi/G(o) and Gs proteins respectively, often co-exist and interact with the phospholipase C-dependent activation of the protein kinase C and the adenylyl cyclase. Activation of the A1 receptor may mimic metabotropic receptor stimulation in activating intracellular Ca2+ mobilization and PKC. A2 receptor mediated cAMP formation is depressed by high intracellular Ca2+ but enhanced by PKC activation. By modulating these metabolic signaling events, adenosine may influence acute cell functions, gene transcription and sustained changes of nerve and glial cells relevant for the development of ischemic damage. The neuroprotective adenosine effect seems to be amplified by treatment with propentofylline, which enhances adenosine release, influences the balance between A1 and A2 receptor mediated actions, depresses the free radical formation in activated microglia and influences astrocyte reactions.


Subject(s)
Adenosine/metabolism , Brain Ischemia/physiopathology , Neuroglia/metabolism , Neurons/metabolism , Adenosine/genetics , Animals , Ion Channels , Ion Transport , Receptors, Purinergic P1/metabolism , Signal Transduction
17.
Gen Pharmacol ; 25(6): 1053-8, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7875526

ABSTRACT

1. Adenosine is an endogenous neuroprotective agent; stimulation of A1 receptors decreases excitatory amino acid neurotransmission and stimulation of A2 receptors inhibits platelet and neutrophil activation and promotes vasodilation. 2. Post-ischemic administration of propentofylline (HWA 285) reduces neuronal damage in gerbils and improves glucose metabolism in all regions of brain in acute stroke patients. 3. Propentofylline inhibits the transport of adenosine into cultured cells and increases extracellular adenosine concentrations in ischemic brain. Thus, enhanced stimulation of adenosine receptors may account for some of the neuroprotective effects of this compound. 4. Propentofylline inhibits free radical production by cultivated microglia cells, stimulates nerve growth factor production and inhibits cAMP-phosphodiesterase activity. These effects may also be important for neuroprotection.


Subject(s)
Adenosine/metabolism , Brain Ischemia/drug therapy , Xanthines , Adenosine/physiology , Animals , Biological Transport/drug effects , Cells, Cultured , Cerebrovascular Circulation/drug effects , Depression, Chemical , Excitatory Amino Acid Antagonists , Gerbillinae , Humans , Receptors, Purinergic P1/drug effects , Xanthines/pharmacology , Xanthines/therapeutic use
18.
Neurosci Lett ; 178(2): 235-8, 1994 Sep 12.
Article in English | MEDLINE | ID: mdl-7824203

ABSTRACT

The effects of the xanthine derivative propentofylline (HWA 285), an inhibitor of adenosine transport, on ischemic brain damage have been evaluated in a model of permanent middle cerebral artery (MCA) occlusion in rats. During the postischemic survival period of 24 h, the animals were subjected to neurological and behavioral observations and then sacrificed to assess the extent of ischemic tissue damage by tetrazolium chloride. Posttreatment with propentofylline (0.01, 0.05 or 0.1 mg/kg/min, continuous i.v. infusion) initiated 15 min following MCA occlusion, produced significant reductions in infarct volumes; the highest being the most effective (reduction by 39%; P < 0.005) and improved the neurological symptoms when compared with an untreated control group. In contrast to other antiischemic agents, such as glutamate receptor antagonists, the drug induced no behavioral disturbances. This study indicates that propentofylline may provide neuroprotective effect against ischemic brain damage following stroke without negative behavioral side effects.


Subject(s)
Brain Ischemia/drug therapy , Cerebral Infarction/drug therapy , Xanthines/pharmacology , Adenosine/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Male , Nervous System/physiopathology , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley
19.
J Med Chem ; 37(19): 3008-15, 1994 Sep 16.
Article in English | MEDLINE | ID: mdl-7932522

ABSTRACT

The preparation and structure-activity relationships of a series of 2-amino-alpha-thienylbenzeneethanamines are described. From this work, (+/-)-2-amino-N-methyl-alpha-(3-methyl-2-thienyl)-benzeneethanamine++ + (3a) and the homologous N-ethyl analog 3b emerged as novel noncompetitive NMDA antagonists with neuroprotective properties. Optical resolution of 3a and X-ray crystallography of (+)3a were performed. The racemate and enantiomers were evaluated for neuroprotective properties in models of ischemia-induced hippocampal damage (gerbil) and cerebral focal ischemia (rat). Pretreatment with 3a, (+)3a, or (-)3a significantly reduced ischemia-induced CA1 hippocampal damage. Posttreatment with 3a afforded a lower degree of neuroprotection. A highly significant reduction in infarct volume was observed with 3a in the cerebral focal ischemia model, with only weak positive effects being displayed by (+)3a. Dose-limiting side effects were associated with all three compounds in this model. In summary, the results demonstrate the utility of noncompetitive NMDA antagonists as neuroprotective agents for ischemia-induced neurodegeneration.


Subject(s)
Aniline Compounds/chemical synthesis , Aniline Compounds/therapeutic use , N-Methylaspartate/antagonists & inhibitors , Thiophenes/chemical synthesis , Thiophenes/therapeutic use , Aniline Compounds/chemistry , Animals , Anti-Anxiety Agents/chemical synthesis , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/therapeutic use , Anticonvulsants/chemical synthesis , Anticonvulsants/therapeutic use , Binding, Competitive , Brain Ischemia/prevention & control , Cerebral Infarction/prevention & control , Crystallography, X-Ray , Disease Models, Animal , Gerbillinae , Hippocampus/blood supply , Hippocampus/drug effects , Isomerism , Male , Mice , Molecular Structure , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Structure-Activity Relationship , Thiophenes/chemistry
20.
Neuroreport ; 5(10): 1193-6, 1994 Jun 02.
Article in English | MEDLINE | ID: mdl-7919163

ABSTRACT

In the gerbil hippocampus activated microglial antigens are intensely stained by cerebrospinal fluid from patients with Alzheimer's disease (AD-CSF), OX18 and the amyloid precursor protein (beta-APP) up to 14 days after ischaemia. Propentosylline (PPF), which facilitates the adenosine A2 receptor action, has been shown to be neuroprotective, to depress O2- radical formation in macrophages and to interfere with the generation of phagocytotic macrophages from cultivated microglial cells. In this report we tested in ischaemic gerbils whether PPF treatment influences the potential neurotoxic properties of microglia. Daily post-treatment with PPF, started 24 h after ischaemia, depressed the immunostaining of activated microglia by AD-CSF, OX18 and APP in the hippocampus. Thus, PPF may protect against microglia-related brain damage.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/immunology , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/immunology , Antigens/immunology , Ischemic Attack, Transient/cerebrospinal fluid , Ischemic Attack, Transient/pathology , Microglia/immunology , Xanthines/pharmacology , Animals , Antigen-Antibody Reactions , Antigens/biosynthesis , Cerebral Ventricles/pathology , Gerbillinae , Hippocampus/pathology , Ischemic Attack, Transient/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...