Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neurotox Res ; 29(3): 432-46, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26739825

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neurotrophic and neuroprotective peptide that has been shown to exert protective effects in different neuronal injuries, such as retinal degenerations. Diabetic retinopathy (DR), the most common complication of diabetes, affects the microvasculature and neuronal architecture of the retina. We have proven earlier that PACAP is also protective in a rat model of DR. In this study, streptozotocin-induced DR was treated with intravitreal PACAP administration in order to further analyze the synaptic structure and proteins of PACAP-treated diabetic retinas, primarily in the vertical information processing pathway. Streptozotocin-treated Wistar rats received intravitreal PACAP injection three times into the right eye 2 weeks after the induction of diabetes. Morphological and molecular biological (qRT-PCR; Western blot) methods were used to analyze retinal synapses (ribbons, conventional) and related structures. Electron microscopic analysis revealed that retinal pigment epithelium, the ribbon synapses and other synaptic profiles suffered alterations in diabetes. However, in PACAP-treated diabetic retinas more bipolar ribbon synapses were found intact in the inner plexiform layer than in DR animals. The ribbon synapse was marked with C-terminal binding protein 2/Bassoon and formed horseshoe-shape ribbons, which were more retained in PACAP-treated diabetic retinas than in DR rats. These results are supported by molecular biological data. The selective degeneration of related structures such as bipolar and ganglion cells could be ameliorated by PACAP treatment. In summary, intravitreal administration of PACAP may have therapeutic potential in streptozotocin-induced DR through maintaining synapse integrity in the vertical pathway.


Subject(s)
Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Neuroprotective Agents/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Retina/metabolism , Retina/ultrastructure , Animals , Diabetic Retinopathy/chemically induced , Diabetic Retinopathy/prevention & control , Male , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Photoreceptor Cells/ultrastructure , Rats , Rats, Wistar , Retina/drug effects , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/ultrastructure , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Streptozocin , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure
2.
Acta Physiol Hung ; 101(1): 67-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24311224

ABSTRACT

Urocortin 2 (Ucn 2) is a corticotrop releasing factor paralog peptide with many physiological functions and it has widespread distribution. There are some data on the cytoprotective effects of Ucn 2, but less is known about its neuro- and retinoprotective actions. We have previously shown that Ucn 2 is protective in ischemia-induced retinal degeneration. The aim of the present study was to examine the protective potential of Ucn 2 in monosodium-glutamate (MSG)-induced retinal degeneration by routine histology and to investigate cell-type specific effects by immunohistochemistry. Rat pups received MSG applied on postnatal days 1, 5 and 9 and Ucn 2 was injected intravitreally into one eye. Retinas were processed for histology and immunocytochemistry after 3 weeks. Immunolabeling was determined for glial fibrillary acidic protein, vesicular glutamate transporter 1, protein kinase Cα, calbindin, parvalbumin and calretinin. Retinal tissue from animals treated with MSG showed severe degeneration compared to normal retinas, but intravitreal Ucn 2 treatment resulted in a retained retinal structure both at histological and neurochemical levels: distinct inner retinal layers and rescued inner retinal cells (different types of amacrine and rod bipolar cells) could be observed. These findings support the neuroprotective function of Ucn 2 in MSG-induced retinal degeneration.


Subject(s)
Neuroprotective Agents/pharmacology , Retina/drug effects , Retinal Degeneration/prevention & control , Sodium Glutamate , Urocortins/pharmacology , Animals , Animals, Newborn , Calbindin 2/metabolism , Calbindins/metabolism , Cytoprotection , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Intravitreal Injections , Neuroprotective Agents/administration & dosage , Parvalbumins/metabolism , Protein Kinase C-alpha/metabolism , Rats , Rats, Wistar , Retina/metabolism , Retina/pathology , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Time Factors , Urocortins/administration & dosage , Vesicular Glutamate Transport Protein 1/metabolism
3.
Neurotox Res ; 25(1): 57-67, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24155155

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide with diverse biological effects. It also occurs and exerts protective effects in sensory organs; however, little is known about its effects in the auditory system. Recently, we have shown that PACAP protects cochlear cells against oxidative-stress-induced apoptosis and homozygous PACAP-deficient animals show stronger expression of Ca(2+)-binding proteins in the hair cells of the inner ear, but there are no data about the consequences of the lack of endogenous PACAP in different ototoxic insults such as aminoglycoside-induced toxicity. In this study, we examined the effect of kanamycin treatment on Ca(2+)-binding protein expression in hair cells of wild-type, heterozygous and homozygous PACAP-deficient mice. We treated 5-day-old mice with kanamycin, and 2 days later, we examined the Ca(2+)-binding protein expression of the hair cells with immunohistochemistry. We found stronger expression of Ca(2+)-binding proteins in the hair cells of control heterozygous and homozygous PACAP-deficient mice compared with wild-type animals. Kanamycin induced a significant increase in Ca(2+)-binding protein expression in wild-type and heterozygous PACAP-deficient mice, but the baseline higher expression in homozygous PACAP-deficient mice did not show further changes after the treatment. Elevated endolymphatic Ca(2+) is deleterious for the cochlear function, against which the high concentration of Ca(2+)-buffers in hair cells may protect. Meanwhile, the increased immunoreactivity of Ca(2+)-binding proteins in the absence of PACAP provide further evidence for the important protective role of PACAP in ototoxicity, but further investigations are necessary to examine the exact role of endogenous PACAP in ototoxic insults.


Subject(s)
Calcium-Binding Proteins/metabolism , Ear, Inner/drug effects , Ear, Inner/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Animals , Calbindin 2/metabolism , Hair Cells, Auditory/metabolism , Heterozygote , Homozygote , Kanamycin/toxicity , Mice , Mice, Knockout , Parvalbumins/metabolism , Protein Synthesis Inhibitors/toxicity
4.
Acta Physiol Hung ; 100(4): 445-56, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24013940

ABSTRACT

UNLABELLED: We hypothesized that stair-jump exercise would induce less muscle damage and greater acute metabolic responses than level-jumps. METHODS: Trained males executed 100 unilateral jumps on stairs with one leg, and at level with the other leg, with two weeks hiatus. Maximal isometric voluntary torque (MVC) and rate of torque development (RTD)in the quadriceps, and unilateral vertical jump height (VJ) were determined in the trained leg at pre-exercise,immediately at post- (IP), 24 h and 48 h after exercise. Serum creatine kinase (CK) level and delayed onset muscle soreness (DOMS) were evaluated at pre-exercise, 24 h and 48 h. Acute lactate and heart rate responses were also measured. RESULTS: Lactate and heart rate at IP increased similarly under the two conditions. CK was elevated and MVC was depressed while RTD and VJ remained unchanged at 24 h in both types of training. MVC recovered at 48 h only after stair-jump exercise. DOMS developed only after level-jumps. Except DOMS, no effects of condition were found in any other variables. CONCLUSIONS: We conclude that vigorous stair-jump exercise highly stresses the aerobic and the anaerobic energy system, and it preserves power and rapid torque generating ability 24 h after exercise. Stair-jump could be one alternative exercise to prevent muscle soreness.


Subject(s)
Isometric Contraction/physiology , Muscle, Skeletal/injuries , Plyometric Exercise/adverse effects , Plyometric Exercise/methods , Creatine Kinase/metabolism , Cross-Over Studies , Energy Metabolism/physiology , Heart Rate/physiology , Humans , Knee Joint/physiology , Leg/physiology , Male , Muscle Fatigue/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Myalgia/metabolism , Myalgia/physiopathology , Young Adult
5.
J Mol Neurosci ; 48(3): 667-73, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22648511

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a pleiotropic and multifunctional neuropeptide having important roles in various physiological processes. Recent trends in PACAP research point to the clinical introduction of PACAP or its analogs/fragments possibly in the near future. Recently, we have shown the presence of PACAP in human plasma, milk, placenta, and follicular fluid samples. However, relatively few data are available on PACAP in human tissues from patients with different disorders. The aim of the present study was to determine, by radioimmunoassay, the tissue level of PACAP38-like immunoreactivity (LI) and PACAP27-LI in different primary non-small cell lung cancer, colon tumor samples, and in cardiac muscle samples from patients suffering from ischemic heart disease and valvular disorders. We also labeled the PAC1 receptors in human cardiac cells. All samples showed significantly higher PACAP38-LI compared with PACAP27-LI. We found significantly lower levels of PACAP38-LI and PACAP27-LI in tumoral and peripheral samples compared with normal healthy tissue in both lung and colon cancers. Further investigations are necessary to describe the exact function of PACAP in oncogenesis. We showed that PACAP38-LI and PACAP27-LI are significantly higher in ischemic heart diseases compared with valvular abnormalities, suggesting that PACAP might play a role in ischemic heart disorders.


Subject(s)
Adenocarcinoma/chemistry , Carcinoma, Non-Small-Cell Lung/chemistry , Colonic Neoplasms/chemistry , Heart Valve Diseases/metabolism , Lung Neoplasms/chemistry , Myocardial Ischemia/metabolism , Myocytes, Cardiac/chemistry , Neoplasm Proteins/analysis , Pituitary Adenylate Cyclase-Activating Polypeptide/analysis , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Colon/chemistry , Colonic Neoplasms/pathology , Heart Valve Diseases/pathology , Humans , Lung/chemistry , Lung Neoplasms/pathology , Myocardial Ischemia/pathology , Myocardium/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Protein Isoforms/analysis , Radioimmunoassay , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/analysis
6.
J Mol Neurosci ; 48(3): 493-500, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22644900

ABSTRACT

The integrity of retinal pigment epithelial cells is critical for photoreceptor survival and vision. Pituitary adenylate cyclase activating polypeptide (PACAP) exerts retinoprotective effects against several types of injuries in vivo, including optic nerve transection, retinal ischemia, excitotoxic injuries, UVA-induced lesion, and diabetic retinopathy. In a recent study, we have proven that PACAP is also protective in oxidative stress-induced injury in human pigment epithelial cells (ARPE-19 cells). The aim of the present study was to investigate the possible mechanisms of this protection. ARPE cells were exposed to a 24-h hydrogen peroxide treatment. Expressions of kinases and apoptotic markers were studied by complex array kits and Western blot. Oxidative stress induced the activation of several apoptotic markers, including Bad, Bax, HIF-1α, several heat shock proteins, TNF-related apoptosis-inducing ligand, and Fas-associated protein with death domain, while PACAP treatment decreased them. The changes in the expression of MAP kinases showed that PACAP activated the protective ERK1/2 and downstream CREB, and decreased the activation of the pro-apoptotic p38MAPK and c-Jun N-terminal kinase, an effect opposite to that observed with only oxidative stress. Furthermore, PACAP increased the activation of the protective Akt pathway. In addition, the effects of oxidative stress on several other signaling molecules were counteracted by PACAP treatment (Chk2, Yes, Lyn, paxillin, p53, PLC, STAT4, RSK). These play a role in cell death, cell cycle, inflammation, adhesion, differentiation and proliferation. In summary, PACAP, acting at several levels, influences the balance between pro- and anti-apoptotic factors in favor of anti-apoptosis, thereby providing protection in oxidative stress-induced injury of human retinal pigment epithelial cells.


Subject(s)
Neuroprotective Agents/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Retinal Pigment Epithelium/drug effects , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured/drug effects , Enzyme Activation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Hydrogen Peroxide/toxicity , Immunoenzyme Techniques , Luminescent Measurements , Oxidative Stress/drug effects , Phosphorylation/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/metabolism , Retinal Pigment Epithelium/cytology
7.
J Mol Neurosci ; 48(3): 501-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22544514

ABSTRACT

Vasoactive intestinal peptide (VIP) is a pleiotropic neuropeptide, acting as a neuromodulator and neuroprotective peptide in the CNS after injuries. We have previously described that pituitary adenylate cyclase-activating polypeptide (PACAP), another member of the same peptide family, is retinoprotective in ischemic lesions. The aim of this study was to investigate the protective potential of VIP in bilateral common carotid artery occlusion (BCCAO)-induced ischemic retinal lesion. Two-month-old rats were subjected to BCCAO and treated with intravitreal VIP injection. Their retinas were processed for histology after 2 weeks of survival. We measured the number of the cells/100 µm of the ganglion cell layer and the thickness of each layer such as the outer nuclear, outer plexiform, inner nuclear, and inner plexiform layers as well as that of the whole retina. We found that treatment with 1,000 pmol VIP, but not 100 pmol VIP, had significant protective effects in BCCAO-injured retina, as shown by the morphometric analysis. Comparing the neuroprotective effects of VIP and PACAP in BCCAO-operated retinas, PACAP was more effective, already protective at 100-pmol doses. Similar to other studies, we found that VIP must be given at least in 10 times more concentration than PACAP to achieve a similar degree of neuroprotection in the retina.


Subject(s)
Carotid Artery, Common , Carotid Stenosis/complications , Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Retinal Degeneration/prevention & control , Vasoactive Intestinal Peptide/therapeutic use , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Intravitreal Injections , Ischemia/etiology , Ischemia/pathology , Male , Models, Neurological , Neuroprotective Agents/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Rats , Rats, Wistar , Retinal Degeneration/etiology , Retinal Degeneration/pathology , Retinal Neurons/drug effects , Retinal Neurons/ultrastructure , Retinal Vessels , Retinitis/drug therapy , Retinitis/etiology , Retinitis/pathology , Time Factors , Vasoactive Intestinal Peptide/administration & dosage , Vasoactive Intestinal Peptide/pharmacology
8.
J Mol Neurosci ; 48(3): 482-92, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22528455

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a widespread neuropeptide with a diverse array of biological functions. Not surprisingly, the lack of endogenous PACAP therefore results in a variety of abnormalities. One of the important effects of PACAP is its neuroprotective and general cytoprotective role. PACAP protects neurons and other tissues against ischemic, toxic, and traumatic lesions. Data obtained from PACAP-deficient mice provide evidence that endogenous PACAP also has protective functions. Mice lacking PACAP are more vulnerable to different in vitro and in vivo insults. The present review summarizes data on the increased sensitivity of PACAP-deficient mice against harmful stimuli. Mice lacking PACAP respond with a higher degree of injury in cerebral ischemia, autoimmune encephalomyelitis, and axonal lesion. Retinal ischemic and excitotoxic injuries also produce increased cell loss in PACAP-deficient mice. In peripheral organs, kidney cell cultures from PACAP-deficient mice are more sensitive to oxidative stress and in vitro hypoxia. In vivo, PACAP-deficient mice have a negative histological outcome and altered cytokine response in kidney and small intestine ischemia/reperfusion injury. Large intestinal inflammation, toxic lesion of the pancreas, and doxorubicin-induced cardiomyopathy are also more severe with a lack of endogenous PACAP. Finally, an increased inflammatory response has been described in subacute endotoxin-induced airway inflammation and in an oxazolone-induced allergic contact dermatitis model. In summary, lack of endogenous PACAP leads to higher vulnerability in a number of injuries in the nervous system and peripheral organs, supporting the hypothesis that PACAP is part of the endogenous cytoprotective machinery.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Stress, Physiological/physiology , Animals , Autoimmune Diseases/physiopathology , Cardiomyopathies/physiopathology , Dermatitis, Allergic Contact/physiopathology , Disease Susceptibility , Homeostasis/physiology , Inflammation/physiopathology , Ischemia/physiopathology , Kidney Diseases/physiopathology , Lung Diseases/physiopathology , Mice , Mice, Knockout , Nervous System Diseases/physiopathology , Neurotoxins/toxicity , Noxae/adverse effects , Pancreatic Diseases/physiopathology , Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Wounds and Injuries/physiopathology
9.
Neurotox Res ; 21(1): 41-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21717232

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuroprotective peptide exerting protective effects in neuronal injuries. We have provided evidence that PACAP is neuroprotective in several models of retinal degeneration in vivo. Our previous studies showed that PACAP treatment ameliorated the damaging effects of chronic hypoperfusion modeled by permanent bilateral carotid artery occlusion. We have also demonstrated in earlier studies that treatment with PACAP antagonists further aggravates retinal lesions. It has been shown that PACAP deficient mice have larger infarct size in cerebral ischemia. The aim of this study was to compare the degree of retinal damage in wild type and PACAP deficient mice in ischemic retinal insult. Mice underwent 10 min of bilateral carotid artery occlusion followed by 2-week reperfusion period. Retinas were then processed for histological analysis. It was found that PACAP deficient mice had significantly greater retinal damage, as shown by the thickness of the whole retina, the morphometric analysis of the individual retinal layers, and the cell numbers in the inner nuclear and ganglion cell layers. Exogenous PACAP administration could partially protect against retinal degeneration in PACAP deficient mice. These results clearly show that endogenous PACAP reacts as a stress-response peptide that is necessary for endogenous protection against different retinal insults.


Subject(s)
Ischemia/etiology , Neuroprotective Agents , Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , Retinal Degeneration/etiology , Retinal Vessels , Animals , Disease Models, Animal , Mice , Mice, Knockout , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Retina/pathology , Retinal Degeneration/pathology
10.
Neurotox Res ; 21(4): 435-44, 2012 May.
Article in English | MEDLINE | ID: mdl-22203609

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a multifunctional neuropeptide with well-known neuroprotective and neurotrophic effects. The involvement of PACAP in sensory processing has also been documented, but little is known about its effects in the auditory system. PACAP and its specific receptor (PAC1) are present in the cochlea and in brain structures involved in auditory pathways. Recently, we have shown that PACAP protects cochlear cells against oxidative stress-induced apoptosis. The endolymphatic Ca(2+) concentration controlled by Ca(2+) buffers of the hair cells is essential for the normal hearing processes. In this study we examined the localization of PAC1 receptor and Ca(2+) buffering proteins (parvalbumin, calretinin, calbindin) in the inner ear of 5-day-old PACAP-deficient mice compared with wild-type mice in order to get a closer insight into the effect of endogenous PACAP in the cochlear function. We did not find differences in the distribution pattern of PAC1 receptors between the two groups, but wild-type animals showed significantly higher PAC1 receptor expression. In contrast, inner and outer hair cells of PACAP-deficient mice showed more pronounced parvalbumin, calbindin, and calretinin immunopositivity compared with wild-type mice. Elevated endolymphatic Ca(2+) is deleterious for cochlear function, while the high concentration of Ca(2+) buffers in hair cells may offer protection. The increased immunoreactivity of Ca(2+) binding proteins in the absence of PACAP provide further evidence the important role of PACAP in the hearing processes.


Subject(s)
Calcium-Binding Proteins/metabolism , Ear, Inner/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Calbindin 2 , Calbindins , Cochlea/metabolism , Ear, Inner/cytology , Hair Cells, Auditory/metabolism , Mice , Mice, Inbred ICR , Mice, Knockout , Parvalbumins/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , S100 Calcium Binding Protein G/metabolism
11.
Ann N Y Acad Sci ; 1200: 128-39, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20633141

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neurotrophic and neuroprotective peptide that has been shown to exert protective effects against different neuronal injuries, such as traumatic brain and spinal cord injury, models of neurodegenerative diseases, and cerebral ischemia. PACAP and its receptors are present in the retina. In this study, we summarize the current knowledge on retinal PACAP with focus on the retinoprotective effects. Results of histological, immunohistochemical, and molecular biological analysis are reviewed. In vitro, PACAP shows protection against glutamate, thapsigargin, anisomycin, and anoxia. In vivo, the protective effects of intravitreal PACAP treatment have been shown in the following models of retinal degeneration in rats: excitotoxic injury induced by glutamate and kainate, ischemic injury, degeneration caused by UV-A light, optic nerve transection, and streptozotocin-induced diabetic retinopathy. Studying the molecular mechanism has revealed that PACAP acts by activating antiapoptotic and inhibiting proapoptotic signaling pathways in the retina in vivo. These studies strongly suggest that PACAP is an excellent candidate retinoprotective agent that could be a potential therapeutic substance in various retinal diseases.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Retina/physiology , Animals , Humans , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Retina/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...