Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(11)2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37298615

ABSTRACT

Tissue plasminogen activator, aiming to restore cerebral blood flow (CBF), has been used for acute ischemic strokes in clinics; however, its narrow therapeutic time window remains a serious concern. To develop novel prophylactic drugs to alleviate cerebral ischemia/reperfusion injuries, ferulic acid derivative 012 (FAD012) was synthesized and showed comparable antioxidant properties to ferulic acid (FA) and probably possesses the potent ability to cross the blood-brain barrier. A more potent cytoprotective effect of FAD012 against H2O2-induced cytotoxicity in PC12 cells was also observed. In vivo toxicity was not observed in rats given a long-term oral administration of FAD012, indicating its good tolerability. A one-week-course oral administration of FAD012 significantly alleviated middle cerebral artery occlusion (MCAO)-induced cerebral ischemia/reperfusion injuries in rats, accompanied by the restoration of CBF and endothelial nitrogen oxide synthetase (eNOS) expression. Treatment with FAD012 significantly restored the cell viability and eNOS expression damaged by H2O2, used to mimic MCAO-triggered oxidative stress, in rat brain microvascular endothelial cells. Our findings suggested that FAD012 protected the viability of vascular endothelium and maintained eNOS expression, ultimately contributing to the restoration of CBF, and may provide a rationale for the development of FAD012 into an effective prophylactic drug for patients at high risk of stroke.


Subject(s)
Brain Ischemia , Neuroprotective Agents , Reperfusion Injury , Animals , Rats , Brain Ischemia/drug therapy , Cerebrovascular Circulation , Endothelial Cells/metabolism , Hydrogen Peroxide/therapeutic use , Infarction, Middle Cerebral Artery/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Tissue Plasminogen Activator/therapeutic use
2.
Molecules ; 27(19)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36235115

ABSTRACT

The cytotoxicity of a trivalent arsenic derivative (arsenite, AsIII) combined with arenobufagin or gamabufotalin was evaluated in human U-87 glioblastoma cells. Synergistic cytotoxicity with upregulated intracellular arsenic levels was observed, when treated with AsIII combined with arenobufagin instead of gamabufotalin. Apoptosis and the activation of caspase-9/-8/-3 were induced by AsIII and further strengthened by arenobufagin. The magnitude of increase in the activities of caspase-9/-3 was much greater than that of caspase-8, suggesting that the intrinsic pathway played a much more important role in the apoptosis. An increase in the number of necrotic cells, enhanced LDH leakage, and intensified G2/M phase arrest were observed. A remarkable increase in the expression level of γH2AX, a DNA damage marker, was induced by AsIII+arenobufagin. Concomitantly, the activation of autophagy was observed, suggesting that autophagic cell death associated with DNA damage was partially attributed to the cytotoxicity of AsIII+arenobufagin. Suppression of Notch signaling was confirmed in the combined regimen-treated cells, suggesting that inactivation of Jagged1/Notch signaling would probably contribute to the synergistic cytotoxic effect of AsIII+arenobufagin. Given that both AsIII and arenobufagin are capable of penetrating into the blood-brain barrier, our findings may provide fundamental insight into the clinical application of the combined regimen for glioblastoma.


Subject(s)
Antineoplastic Agents , Arsenic , Arsenites , Bufanolides , Glioblastoma , Antineoplastic Agents/pharmacology , Apoptosis , Arsenic/metabolism , Arsenites/pharmacology , Bufanolides/pharmacology , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans
3.
Int J Pharm ; 594: 120046, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33309832

ABSTRACT

Acetaminophen, a central antipyretic and analgesic drug, is one of the most commonly used drugs among individuals of all ages throughout the world. This study pharmacokinetically and pharmacodynamically investigated the transport of acetaminophen to the central nervous system and systemic circulation after intranasal (i.n.) administration, and evaluated the potential of a transnasal acetaminophen formulation in comparison to other routes of administration. Direct transport to the brain and the pharmacological effect after the i.n. administration of acetaminophen with polyvinylpyrrolidone (PVP; a mucoadhesive agent) and poly-l-arginine (PLA; an absorption enhancer) were investigated to improve retention of the dosage solution in the olfactory epithelium region and enhance the transfer of acetaminophen to the brain. The transport of acetaminophen to the brain was rapid, and the concentration in the brain, especially the olfactory bulb, was higher after i.n. administration, resulting in a greater antipyretic effect in comparison to other routes of administration. The delivery system using PVP and PLA produced a high and prolonged antipyretic effect by enhancing the transfer of acetaminophen to the brain through suppression of the transfer to systemic circulation. Thus, this transnasal drug delivery system using PVP and PLA may be a promising method for transporting acetaminophen to the brain.


Subject(s)
Acetaminophen , Central Nervous System , Administration, Intranasal , Brain , Drug Delivery Systems , Humans
4.
Toxicol Sci ; 157(2): 421-428, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28402547

ABSTRACT

The brain level of perfluorododecanoic acid (PFDoA) was compared with those of perfluorooctanoic acid (PFOA) and perfluorodecanoic acid (PFDA) in rats 9 days after a single oral dose (50 mg/kg). The PFDoA level in the brain was 44.0 ± 2.0 µg/g, which was higher than that in the serum (24.4 ± 1.0 µg/ml). In contrast, the concentrations of PFOA and PFDA in the brain were low (<0.8 and 4.7 ± 0.4 µg/g, respectively), and less than one-tenth of those in the serum. Next, to investigate the effects on brain function, the cognitive function alterations of PFOA, PFDA, and PFDoA were estimated by the novel object recognition test 5-6 days after dosing. A significant decrease in the discrimination index was observed in PFDoA-treated rats while no significant alteration was observed in PFDA- and PFOA-treated rats. The effects of PFDoA were further assessed by other behavioral tests. PFDoA-associated alteration was observed in the elevated-plus maze test, but not in the Y-maze test, open-field test, and forced swim test. A decrease in the discrimination index of the novel object recognition test was dependent on the PFDoA dose and the PFDoA concentration in the brain. PFDoA concentration in the brain was 28.6 ± 2.6 µg/g 30 days after dosing, and a decrease in discrimination index was observed. Taken together, these results suggest that PFDoA distributes in the brain easier than PFOA and PFDA and causes cognitive deficit.


Subject(s)
Brain/drug effects , Cognitive Dysfunction/chemically induced , Environmental Pollutants/toxicity , Lauric Acids/toxicity , Animals , Behavior, Animal/drug effects , Brain/metabolism , Cognitive Dysfunction/metabolism , Environmental Pollutants/pharmacokinetics , Fluorocarbons , Lauric Acids/pharmacokinetics , Male , Maze Learning/drug effects , Rats, Wistar , Tissue Distribution
5.
Int J Mol Sci ; 18(3)2017 Mar 03.
Article in English | MEDLINE | ID: mdl-28273833

ABSTRACT

Ferulic acid (FA), a phenolic phytochemical, has been reported to exert antioxidative and neuroprotective effects. In this study, we investigated the protective effects of FA against the dysfunction of the swallowing reflex induced by ligation of bilateral common carotid arteries (2VO) in rats. In 2VO rats, topical administration of water or citric acid to the pharyngolaryngeal region evoked a diminished number of swallowing events with prolonged latency compared to sham-operated control rats. 2VO rats had an increased level of superoxide anion radical, and decreased dopamine and tyrosine hydroxylase enzyme levels in the striatum, suggesting that 2VO augmented cerebral oxidative stress and impaired the striatal dopaminergic system. Furthermore, substance P (SP) expression in the laryngopharyngeal mucosa, which is believed to be positively regulated by dopaminergic signaling in the basal ganglia, was decreased in 2VO rats. Oral treatment with FA (30 mg/kg) for 3 weeks (from one week before 2VO to two weeks after) improved the swallowing reflex and maintained levels of striatal dopamine and laryngopharyngeal SP expression in 2VO rats. These results suggest that FA maintains the swallowing reflex by protecting the dopamine-SP system against ischemia-induced oxidative damage in 2VO rats.


Subject(s)
Brain Ischemia/etiology , Brain Ischemia/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Coumaric Acids/pharmacology , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Cerebrovascular Circulation/drug effects , Corpus Striatum/blood supply , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/biosynthesis , Dopaminergic Neurons/metabolism , Gene Expression , Male , Oxidative Stress/drug effects , Rats , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
6.
Biomed Res Int ; 2015: 189292, 2015.
Article in English | MEDLINE | ID: mdl-26665003

ABSTRACT

Diabetes mellitus is known to exacerbate acute cerebral ischemic injury. Previous studies have demonstrated that infarction volumes caused by transient cerebral ischemia were greater in diabetic rats than in nondiabetic rats. Tumor necrosis factor-α (TNF-α) is a proinflammatory protein produced in the brain in response to cerebral ischemia that promotes apoptosis. Etanercept (ETN), a recombinant TNF receptor (p75)-Fc fusion protein, competitively inhibits TNF-α. Therefore, we evaluated the neuroprotective effects of chronic or acute treatment with ETN on cerebral injury caused by middle cerebral artery occlusion/reperfusion (MCAO/Re) in rats with streptozotocin-induced diabetes. Furthermore, we evaluated the effects of ETN against the apoptosis and myeloperoxidase activity. Single administration of ETN before MCAO significantly suppressed exacerbation of cerebral damage in nondiabetic rats, as assessed by infarct volume. In contrast, the diabetic state markedly aggravated MCAO/Re-induced cerebral damage despite ETN treatment within 24 h before MCAO. However, the damage was improved by repeated administration of ETN at 900 µg/kg/daily in rats in an induced diabetic state. These results suggested that repeated administration of ETN can prevent exacerbation of cerebral ischemic injury in the diabetic state and is mainly attributed to anti-inflammatory effects.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Etanercept/administration & dosage , Inflammation/drug therapy , Ischemic Attack, Transient/drug therapy , Animals , Apoptosis/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/physiopathology , Inflammation/genetics , Inflammation/pathology , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/physiopathology , Neuroprotective Agents , Rats , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
7.
Article in English | MEDLINE | ID: mdl-25945116

ABSTRACT

Type 2 diabetes mellitus has been known to increase systemic oxidative stress by chronic hyperglycemia and visceral obesity and aggravate cerebral ischemic injury. On the basis of our previous study regarding a water-soluble extract from the culture medium of Ganoderma lucidum mycelia (designed as MAK), which exerts antioxidative and neuroprotective effects, the present study was conducted to evaluate the preventive effects of MAK on apoptosis and necroptosis (a programmed necrosis) induced by hypoxia/ischemia (H/I) in type 2 diabetic KKAy mice. H/I was induced by a combination of unilateral common carotid artery ligation with hypoxia (8% O2 for 20 min) and subsequent reoxygenation. Pretreatment with MAK (1 g/kg, p.o.) for a week significantly reduced H/I-induced neurological deficits and brain infarction volume assessed at 24 h of reoxygenation. Histochemical analysis showed that MAK significantly suppressed superoxide production, neuronal cell death, and vacuolation in the ischemic penumbra, which was accompanied by a decrease in the numbers of TUNEL- or cleaved caspase-3-positive cells. Furthermore, MAK decreased the expression of receptor-interacting protein kinase 3 mRNA and protein, a key molecule for necroptosis. These results suggest that MAK confers resistance to apoptotic and necroptotic cell death and relieves H/I-induced cerebral ischemic injury in type 2 diabetic mice.

8.
Nutrients ; 6(4): 1554-77, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24739976

ABSTRACT

Diabetes mellitus is known to exacerbate cerebral ischemic injury. In the present study, we investigated antiapoptotic and anti-inflammatory effects of oral supplementation of ascorbic acid (AA) on cerebral injury caused by middle cerebral artery occlusion and reperfusion (MCAO/Re) in rats with streptozotocin-induced diabetes. We also evaluated the effects of AA on expression of sodium-dependent vitamin C transporter 2 (SVCT2) and glucose transporter 1 (GLUT1) after MCAO/Re in the brain. The diabetic state markedly aggravated MCAO/Re-induced cerebral damage, as assessed by infarct volume and edema. Pretreatment with AA (100 mg/kg, p.o.) for two weeks significantly suppressed the exacerbation of damage in the brain of diabetic rats. AA also suppressed the production of superoxide radical, activation of caspase-3, and expression of proinflammatory cytokines (tumor necrosis factor-α and interleukin-1ß) in the ischemic penumbra. Immunohistochemical staining revealed that expression of SVCT2 was upregulated primarily in neurons and capillary endothelial cells after MCAO/Re in the nondiabetic cortex, accompanied by an increase in total AA (AA + dehydroascorbic acid) in the tissue, and that these responses were suppressed in the diabetic rats. AA supplementation to the diabetic rats restored these responses to the levels of the nondiabetic rats. Furthermore, AA markedly upregulated the basal expression of GLUT1 in endothelial cells of nondiabetic and diabetic cortex, which did not affect total AA levels in the cortex. These results suggest that daily intake of AA attenuates the exacerbation of cerebral ischemic injury in a diabetic state, which may be attributed to anti-apoptotic and anti-inflammatory effects via the improvement of augmented oxidative stress in the brain. AA supplementation may protect endothelial function against the exacerbated ischemic oxidative injury in the diabetic state and improve AA transport through SVCT2 in the cortex.


Subject(s)
Ascorbic Acid/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Glucose Transporter Type 1/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Neurons/drug effects , Sodium-Coupled Vitamin C Transporters/metabolism , Administration, Oral , Animals , Apoptosis/drug effects , Brain/drug effects , Caspase 3 , Dietary Supplements , Endothelial Cells/drug effects , Glucose Transporter Type 1/genetics , Infarction, Middle Cerebral Artery/pathology , Interleukin-1beta/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Sodium-Coupled Vitamin C Transporters/genetics , Streptozocin , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
9.
BMC Complement Altern Med ; 13: 370, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24369991

ABSTRACT

BACKGROUND: Ganoderma lucidum is a popular medicinal mushroom used for promoting health and longevity in Asian countries. Previously, we reported that a water-soluble extract from a culture medium of Ganoderma lucidum mycelia (MAK) exerts antioxidative and cerebroprotective effects against ischemia-reperfusion injury in vivo. Here, we evaluated the antidepressant and anxiolytic activities of MAK in rats. METHODS: MAK (0.3 or 1 g/kg, p.o.) was administered in the experimental animals 60 min before the forced swimming, open-field, elevated plus-maze, contextual fear-conditioning, and head twitch tests. Additionally, the mechanisms involved in the antidepressant-like action of MAK were investigated by the serotonin precursor 5-hydroxy-L-tryptophan (5-HTP)- or 5-HT2A agonist (±)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane hydrochloride (DOI)-induced head twitch responses. RESULTS: Treatment with MAK (1 g/kg) exhibited antidepressant-like effects in the forced swimming test, attenuated freezing behavior in the contextual fear-conditioning test, and decreased the number of head twitches induced by DOI, but not with 5-HTP. No significant response was observed in locomotion or anxiety-like behavior, when the animals were evaluated in the open-field or elevated plus-maze test, respectively. CONCLUSIONS: These data suggest that MAK has antidepressant-like potential, which is most likely due to the antagonism of 5-HT2A receptors, and possesses anxiolytic-like effects toward memory-dependent and/or stress-induced anxiety in rats.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Plant Extracts/pharmacology , Reishi/chemistry , 5-Hydroxytryptophan/toxicity , Analysis of Variance , Animals , Antidepressive Agents/chemistry , Antidepressive Agents/therapeutic use , Culture Media, Conditioned , Fear/drug effects , Male , Mycelium/metabolism , Plant Extracts/chemistry , Rats , Rats, Sprague-Dawley , Reishi/metabolism , Stress, Physiological , Stress, Psychological , Tics/chemically induced , Tics/drug therapy , Tics/physiopathology
10.
Eur J Pharmacol ; 696(1-3): 89-95, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23036373

ABSTRACT

The amygdala and the medial prefrontal cortex (mPFC) are crucial brain structures for anxiety, and it is speculated that the serotonergic neural system in these structures has an important role in regulating anxiety. In our previous study, we indicated that local injections of selective serotonin reuptake inhibitor into the amygdala attenuated anxiety-related behaviors in conditioned fear in rats. In the present study, we investigated the effects of serotonergic terminal lesions in the amygdala and in mPFC induced by local injection of 5,7-dihydroxytryptamine (5,7-DHT), on anxiety-related behaviors in conditioned fear and the elevated plus-maze test in rats. A 5,7-DHT lesion in the amygdala attenuated memory-dependent fear assessed by conditioned fear, but enhanced memory-independent fear assessed by the elevated plus-maze test. These results suggest that the role of the amygdalar serotonergic system in fear is different between memory-dependent and independent fear and, in particular, it is paradoxical that an amygdalar serotonergic lesion exerts a similar effect on memory-dependent fear to SSRI. Moreover, a serotonergic lesion in the amygdala enhanced the retrieval of extinction memory in conditioned fear; however, a serotonergic lesion in mPFC did not bring about any behavioral changes.


Subject(s)
5,7-Dihydroxytryptamine/pharmacology , Amygdala/drug effects , Behavior, Animal/drug effects , Fear/drug effects , Serotonin Agents/pharmacology , Amygdala/metabolism , Animals , Conditioning, Classical , Male , Memory/drug effects , Rats , Rats, Wistar , Serotonin/metabolism
11.
Psychopharmacology (Berl) ; 214(1): 329-37, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20714708

ABSTRACT

RATIONALE: Traumatic events in early life are associated with an increased risk of psychiatric diseases in adulthood. 5-hydroxytryptamine (5-HT)(1A) receptors play a pivotal role in the 5-HTergic mechanisms associated with the etiology of stress-related disorders. OBJECTIVE: The goal of the present study was to investigate whether juvenile stress influences emotional control via postsynaptic 5-HT(1A) receptor in the hippocampus and amygdala using contextual fear conditioning test in adult rats. METHODS: The rats were subjected to aversive footshock (FS) during the third week of the postnatal period (3wFS group). During the postadolescent period (10-14 weeks postnatal), experiments were performed. RESULTS: The systemic administration of the 5-HT(1A) receptor agonist R-(+)-8-hydroxy-2-(di-n-propylamino) tetralin (8-OH-DPAT) (0.2 mg/kg, i.p.) attenuated the freezing behavior in the non-FS group, but not in the 3wFS group. The bilateral local injection of 8-OH-DPAT (1 µg/side) into the amygdala decreased the freezing behavior in the non-FS group and the 3wFS group. However, the local injection of 8-OH-DPAT (1 µg/side) into the hippocampus decreased the freezing behavior in the non-FS group, but not in the 3wFS group. In a 5-HT(1A) receptor binding study, the Bmax of the 3wFS group decreased in the dorsal hippocampus, but not the amygdala in comparison with the non-FS group. CONCLUSIONS: The juvenile stress attenuated the hippocampal postsynaptic 5-HT(1A) receptor function in context-dependent conditioned fear.


Subject(s)
Hippocampus/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Stress, Psychological , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Age Factors , Animals , Disease Models, Animal , Fear , Freezing Reaction, Cataleptic , Male , Protein Binding , Rats , Rats, Wistar , Serotonin 5-HT1 Receptor Agonists/pharmacology
12.
Eur J Pharmacol ; 615(1-3): 76-82, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19470384

ABSTRACT

Traumatic events in early life are associated with an increased risk of psychiatric diseases in adulthood. 5-hydroxytryptamine (5-HT)(1A) receptors are known to play a pivotal role in the 5-HTergic mechanisms associated with the etiology of stress-related disorders. The goal of the present study was to investigate whether early postnatal stress influences 5-HT(1A) receptor function in the medial prefrontal cortex in adult rats. Rats were subjected to aversive foot shock (FS) during the third week of the postnatal period (3wFS group). During the postadolescent period (10-14 weeks postnatal), immunohistochemical experiments were carried out to investigate c-Fos expression following the administration of R-(+)-8-hydroxy-2-(di-n-propylamino) tetralin (8-OH-DPAT), a 5-HT(1A) receptor agonist. In the 3wFS group, the 8-OH-DPAT-induced c-Fos expression in the medial prefrontal cortex was significantly attenuated compared to that in the non-FS control group. A dual immunofluorescence study revealed that a small proportion of c-Fos positive cells co-express parvalbumin, and a relatively large proportion of c-Fos positive cells co-express glutaminase, suggesting that most c-Fos positive cells are glutamatergic neurons. We found that local perfusion of 8-OH-DPAT via a dialysis probe decreased extracellular 5-HT levels in the medial prefrontal cortex of the non-FS group, but not in the 3wFS group. However, the levels of 8-OH-DPAT-induced 5-HT syndrome were not significantly different between the non-FS and 3wFS groups. Therefore, aversive stress in the third week of the postnatal period attenuates 5-HT(1A) receptor function in the medial prefrontal cortex in adulthood and produces feedback inhibition of the raphe nuclei via postsynaptic 5-HT(1A) receptors.


Subject(s)
Aging/metabolism , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Serotonin/metabolism , Stress, Psychological/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Animals, Newborn , Behavior, Animal/drug effects , Disease Models, Animal , Electroshock , Extracellular Space/metabolism , Feedback, Physiological , Male , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Serotonin 5-HT1 Receptor Agonists , Stress, Psychological/physiopathology
13.
Microsc Res Tech ; 60(5): 491-502, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12619125

ABSTRACT

The periodontal ligament, located between the cementum and the alveolar bone, has a width ranging from 0.15 to 0.38 mm. Regeneration and homeostasis of the periodontal ligament are highly significant functions in relation to periodontal therapy, tooth transplantation or replantation, and orthodontic tooth movement. The purpose of this review is to discuss the regulatory mechanisms of regenerative and homeostatic functions in the periodontal ligament based on currently published studies and also on our own experimental data. We consider the capability of the ligament tissue to promote or to suppress calcification in connection with bone and cementum formation and the maintenance of the periodontal ligament space. Also discussed are the involvement of the periodontal ligament tissue in the regenerative ability, cell proliferation, growth and differentiation factors, extracellular matrix proteins, homeostatic phenomena, function of Malassez epithelial rests, tooth movement, or occlusal loading. Regulatory mechanisms for regeneration and homeostasis of the periodontal ligament are hypothetically proposed.


Subject(s)
Periodontal Ligament/physiology , Regeneration , Animals , Dogs , Gene Expression Regulation, Developmental , Homeostasis , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...