Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
eNeuro ; 8(2)2021.
Article in English | MEDLINE | ID: mdl-33632814

ABSTRACT

Secondary damage after spinal cord injury (SCI) occurs because of a sequence of events after the initial injury, including exacerbated inflammation that contributes to increased lesion size and poor locomotor recovery. Thus, mitigating secondary damage is critical to preserve neural tissue and improve neurologic outcome. In this work, we examined the therapeutic potential of a novel antisense oligonucleotide (ASO) with special chemical modifications [2'-deoxy-2-fluoro-D-arabinonucleic acid (FANA) ASO] for specifically inhibiting an inflammatory molecule in the injured spinal cord. The chemokine CCL3 plays a complex role in the activation and attraction of immune cells and is upregulated in the injured tissue after SCI. We used specific FANA ASO to inhibit CCL3 in a contusive mouse model of murine SCI. Our results show that self-delivering FANA ASO molecules targeting the chemokine CCL3 penetrate the spinal cord lesion site and suppress the expression of CCL3 transcripts. Furthermore, they reduce other proinflammatory cytokines such as tumor necrosis factor (TNF) and interleukin (IL)-1ß after SCI. In summary, we demonstrate for the first time the potential of FANA ASO molecules to penetrate the spinal cord lesion site to specifically inhibit CCL3, reducing proinflammatory cytokines and improve functional recovery after SCI. This novel approach may be used in new treatment strategies for SCI and other pathologic conditions of the CNS.


Subject(s)
Oligonucleotides , Spinal Cord Injuries , Animals , Disease Models, Animal , Inflammation , Mice , Recovery of Function , Spinal Cord , Spinal Cord Injuries/drug therapy
3.
J Neuroinflammation ; 17(1): 362, 2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33246483

ABSTRACT

BACKGROUND: Secondary damage after spinal cord injury (SCI) is characterized by a cascade of events including hemorrhage, apoptosis, oxidative stress, and inflammation which increase the lesion size which can influence the functional impairment. Thus, identifying specific mechanisms attributed to secondary injury is critical in minimizing tissue damage and improving neurological outcome. In this work, we are investigating the role of CCL3 (macrophage inflammatory protein 1-α, MIP-1α), a chemokine involved in the recruitment of inflammatory cells, which plays an important role in inflammatory conditions of the central and peripheral nervous system. METHODS: A mouse model of lower thoracic (T11) spinal cord contusion injury was used. We assessed expression levels of CCL3 and its receptors on the mRNA and protein level and analyzed changes in locomotor recovery and the inflammatory response in the injured spinal cord of wild-type and CCL3-/- mice. RESULTS: The expression of CCL3 and its receptors was increased after thoracic contusion SCI in mice. We then examined the role of CCL3 after SCI and its direct influence on the inflammatory response, locomotor recovery and lesion size using CCL3-/- mice. CCL3-/- mice showed mild but significant improvement of locomotor recovery, a smaller lesion size and reduced neuronal damage compared to wild-type controls. In addition, neutrophil numbers as well as the pro-inflammatory cytokines and chemokines, known to play a deleterious role after SCI, were markedly reduced in the absence of CCL3. CONCLUSION: We have identified CCL3 as a potential target to modulate the inflammatory response and secondary damage after SCI. Collectively, this study shows that CCL3 contributes to progressive tissue damage and functional impairment during secondary injury after SCI.


Subject(s)
Chemokine CCL3/immunology , Spinal Cord Injuries/pathology , Animals , Chemokine CCL3/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recovery of Function , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism
4.
Neurosci Lett ; 660: 51-56, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28899787

ABSTRACT

Ryanodine receptors (RyRs) are highly conductive intracellular Ca2+ release channels and are widely expressed in many tissues, including the central nervous system. RyRs have been implicated in intracellular Ca2+ overload which can drive secondary damage following traumatic injury to the spinal cord (SCI), but the spatiotemporal expression of the three isoforms of RyRs (RyR1-3) after SCI remains unknown. Here, we analyzed the gene and protein expression of RyR isoforms in the murine lumbar dorsal root ganglion (DRG) and the spinal cord lesion site at 1, 2 and 7 d after a mild contusion SCI. Quantitative RT PCR analysis revealed that RyR3 was significantly increased in lumbar DRGs and at the lesion site at 1 and 2 d post contusion compared to sham (laminectomy only) controls. Additionally, RyR2 expression was increased at 1 d post injury within the lesion site. RyR2 and -3 protein expression was localized to lumbar DRG neurons and their spinal projections within the lesion site acutely after SCI. In contrast, RyR1 expression within the DRG and lesion site remained unaltered following trauma. Our study shows that SCI initiates acute differential expression of RyR isoforms in DRG and spinal cord.


Subject(s)
Ryanodine Receptor Calcium Release Channel/genetics , Spinal Cord Injuries/metabolism , Animals , Ganglia, Spinal/metabolism , Gene Expression , Mice , Mice, Transgenic , Protein Isoforms/genetics
5.
Neurobiol Dis ; 106: 235-243, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28709993

ABSTRACT

Severed CNS axons often retract or dieback away from the injury site and fail to regenerate. The precise mechanisms underlying acute axonal dieback and secondary axonal degeneration remain poorly understood. Here we investigate the role of Ca2+ store mediated intra-axonal Ca2+ release in acute axonal dieback and secondary axonal degeneration. To differentiate between primary (directly transected) and "bystander" axonal injury (axons spared by the initial injury but then succumb to secondary degeneration) in real-time we use our previously published highly focal laser-induced spinal cord injury (LiSCI) ex vivo model. Ascending spinal cord dorsal column axons that express YFP were severed using an 800 nm laser pulse while being imaged continuously using two-photon excitation microscopy. We inhibited two major intra-axonal Ca2+ store channels, ryanodine receptors (RyR) and IP3R, with ryanodine or 2-APB, respectively, to individually determine their role in axonal dieback and secondary axonal degeneration. Each antagonist was dissolved in artificial CSF and applied 1h post-injury alone or in combination, and continuously perfused for the remainder of the imaging session. Initially following LiSCI, transected axons retracted equal distances both distal and proximal to the lesion. However, by 4h after injury, the distal axonal segments that are destined for Wallerian degeneration had significantly retracted further than their proximal counterparts. We also found that targeting either RyR or IP3R using pharmacological and genetic approaches significantly reduced proximal axonal dieback and "bystander" secondary degeneration of axons compared to vehicle controls at 6h post-injury. Combined treatment effects on secondary axonal degeneration were similar to either drug in isolation. Together, these results suggest that intra-axonal Ca2+ store mediated Ca2+ release through RyR or IP3R contributes to secondary axonal degeneration following SCI.


Subject(s)
Axons/metabolism , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Intracellular Space/metabolism , Nerve Degeneration/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Axons/drug effects , Axons/pathology , Calcium Channel Blockers/pharmacology , Cations, Divalent/metabolism , Gene Knockdown Techniques , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/genetics , Intracellular Space/drug effects , Lasers , Mice, Transgenic , Nerve Degeneration/pathology , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , RNA, Messenger/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Tissue Culture Techniques
6.
Exp Neurol ; 294: 1-11, 2017 08.
Article in English | MEDLINE | ID: mdl-28445714

ABSTRACT

Microglia/macrophage activation and recruitment following spinal cord injury (SCI) is associated with both detrimental and reparative functions. Stimulation of the innate immune receptor Toll-like receptor-2 (TLR2) has shown to be beneficial following SCI, and it increases axonal regeneration following optic nerve crush. However, the mechanism(s) remain unclear. As microglia express high levels of TLR2, we hypothesized that modulating the microglial response to injury using a specific TLR2 agonist, Pam3CSK4, would prevent secondary-mediated white matter degeneration following SCI. To test this hypothesis, we documented acute changes in microglia, axons, and oligodendroglia over time using two-photon excitation and an ex vivo laser-induced SCI (LiSCI) model. We utilized double transgenic mice that express GFP in either microglia or oligodendroglia, and YFP in axons, and we applied the lipophilic fluorescent dye (Nile Red) to visualize myelin. We found that treatment with Pam3CSK4 initiated one hour after injury induced a significant increase in the extent and timing of the microglial response to injury compared to vehicle controls. This enhanced response was observed 2 to 4h following SCI and was most prominent in areas closer to the ablation site. In addition, Pam3CSK4 treatment significantly reduced axonal dieback rostral and caudal to the ablation at 6h post-SCI. This protective effect of Pam3CSK4 was also mirrored when assessing secondary bystander axonal damage (i.e., axons spared by the primary injury that then succumb to secondary degeneration), and when assessing the survival of oligodendroglia. Following these imaging experiments, custom microarray analysis of the ex vivo spinal cord preparations revealed that Pam3CSK4-treatment induced an alternative (mixed M1:M2) microglial activation profile. In summary, our data suggest that by providing a second "sterile" activation signal to microglia through TLR2/TLR1 signaling, the microglial response to injury can be modulated in situ and is highly neuroprotective.


Subject(s)
Gene Expression Regulation/drug effects , Lipopeptides/pharmacology , Lipopeptides/therapeutic use , Nerve Degeneration/drug therapy , Spinal Cord Injuries/drug therapy , 2',3'-Cyclic-Nucleotide Phosphodiesterases/genetics , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Animals , Axons/drug effects , Axons/pathology , CX3C Chemokine Receptor 1 , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Inflammation/drug therapy , Inflammation/etiology , Inflammation/metabolism , Laser Therapy/adverse effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macrophage Activation , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Microglia/drug effects , Nerve Degeneration/etiology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, Cell Surface/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Immunologic , Spinal Cord Injuries/complications , Spinal Cord Injuries/etiology , Spinal Cord Injuries/pathology , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism
7.
PLoS One ; 10(4): e0124510, 2015.
Article in English | MEDLINE | ID: mdl-25915660

ABSTRACT

Multiple sclerosis (MS) is characterized by inflammatory demyelination and deposition of fibrinogen in the central nervous system (CNS). Elevated levels of a critical inhibitor of the mammalian fibrinolitic system, plasminogen activator inhibitor 1 (PAI-1) have been demonstrated in human and animal models of MS. In experimental studies that resemble neuroinflammatory disease, PAI-1 deficient mice display preserved neurological structure and function compared to wild type mice, suggesting a link between the fibrinolytic pathway and MS. We previously identified a series of PAI-1 inhibitors on the basis of the 3-dimensional structure of PAI-1 and on virtual screening. These compounds have been reported to provide a number of in vitro and in vivo benefits but none was tested in CNS disease models because of their limited capacity to penetrate the blood-brain barrier (BBB). The existing candidates were therefore optimized to obtain CNS-penetrant compounds. We performed an in vitro screening using a model of BBB and were able to identify a novel, low molecular PAI-1 inhibitor, TM5484, with the highest penetration ratio among all other candidates. Next, we tested the effects on inflammation and demyelination in an experimental allergic encephalomyelitis mice model. Results were compared to either fingolimod or 6α-methylprednisolone. Oral administration of TM5484 from the onset of signs, ameliorates paralysis, attenuated demyelination, and axonal degeneration in the spinal cord of mice. Furthermore, it modulated the expression of brain-derived neurotrophic factor, which plays a protective role in neurons against various pathological insults, and choline acetyltransferase, a marker of neuronal density. Taken together, these results demonstrate the potential benefits of a novel PAI-1 inhibitor, TM5484, in the treatment of MS.


Subject(s)
Demyelinating Diseases/pathology , Multiple Sclerosis/pathology , Neurons/drug effects , Neurons/pathology , Plasminogen Activator Inhibitor 1/pharmacology , Animals , Axons , Biological Availability , Brain-Derived Neurotrophic Factor/metabolism , Choline O-Acetyltransferase/metabolism , Collagen Type IV/metabolism , Demyelinating Diseases/drug therapy , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Multiple Sclerosis/drug therapy , Paralysis/drug therapy , Paralysis/etiology , Permeability , Psychomotor Performance/drug effects
8.
Curr Diabetes Rev ; 11(2): 107-15, 2015.
Article in English | MEDLINE | ID: mdl-25732031

ABSTRACT

Excessive dietary intake of fat is strongly involved in the development of type 2 diabetes (T2D). Free fatty acids (FFAs), which are provided from dietary fat, are not only important nutrients, but also act as signaling molecules and stimulate key biological functions. Recent physiological and pharmacological studies have shown that several G-protein coupled receptors, such as FFAR1-4, are receptors for FFAs. FFAR1 and FFAR4 are activated by medium- and long-chain fatty acids, whereas FFAR2 and FFAR3 are activated by short-chain fatty acids (SCFAs). These FFA receptors (FFARs) mediate various physiological functions, depending on the carbon chain length of the FFAs and the ligand specificity of the FFARs. Functional analyses have revealed that FFARs mediate important metabolic functions, such as peptide hormone secretion and inflammation, and thereby contribute to energy homeostasis. Since imbalances in energy homeostasis lead to metabolic disorders, such as obesity and T2D, FFARs are considered to be key therapeutic targets in these diseases. In particular, recent studies have shown that the administration of selective agonists of FFAR1 and FFAR4 improved glucose metabolism and ameliorated systemic metabolic disorders. Furthermore, the biological functions of SCFAs in anti-inflammation and energy metabolism are linked with the activation of FFAR2 and FFAR3. Hence, in this review, we summarize the physiological functions of FFARs and discuss the potential of selective ligands of FFARs for development as drugs to treat metabolic disorders, such as T2D and obesity.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Fatty Acids, Nonesterified/classification , Fatty Acids, Nonesterified/physiology , Hypoglycemic Agents/therapeutic use , Obesity/drug therapy , Drug Delivery Systems , Energy Metabolism , Fatty Acids, Nonesterified/agonists , Homeostasis , Humans
10.
Curr Hypertens Rev ; 9(2): 93-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23971690

ABSTRACT

Recent studies have suggested that blood-brain barrier (BBB) abnormalities are present from an early stage in patients exhibiting mild symptoms of cognitive impairment during the development of hypertension. There is also growing body of evidence suggesting the potential role of the renin-angiotensin system (RAS) in the pathogenesis of small-vessel disease and cognitive impairment. However, the specific contribution of the RAS to BBB disruption and cognitive impairment remains unclear. We found a significant leakage from brain microvessels in the hippocampus and impaired cognitive functions in angiotensin II (AngII)-infused hypertensive mice, which were associated with increased brain AngII levels. These changes were not observed in AngII-infused AT1a receptor (-/-) mice. We also observed that Dahl salt-sensitive hypertensive rats exhibited hypertension, leakage from brain microvessels in the hippocampus, and impaired cognitive function. In these animals, treatment with an AngII receptor blocker, olmesartan, did not alter blood pressure, but markedly ameliorated leakage from brain microvessels and restored the cognitive decline. These data support the hypothesis that RAS inhibition attenuates cognitive impairment by reducing BBB injury, which is independent of blood pressure changes.


Subject(s)
Angiotensin Receptor Antagonists/pharmacology , Blood-Brain Barrier/drug effects , Cognition Disorders/prevention & control , Hypertension/physiopathology , Renin-Angiotensin System/physiology , Animals , Brain/blood supply , Capillary Permeability , Mice , Rats , Rats, Inbred Dahl
11.
J Pharmacol Sci ; 122(2): 103-8, 2013.
Article in English | MEDLINE | ID: mdl-23698111

ABSTRACT

Angiotensin II (Ang II) infusion into rats elevates local angiotensin II levels through an AT1 receptor-dependent pathway in the kidney. We examined whether treatment with an angiotensin-converting enzyme (ACE) inhibitor, temocapril, or an AT1-receptor blocker, olmesartan, prevented elevation of Ang II levels in the kidney of angiotensin I (Ang I)-infused rats. Rats were infused with Ang I (100 ng/min) and treated with temocapril (30 mg/kg per day, n = 10) or olmesartan (10 mg/kg per day, n = 9) for 4 weeks. Ang I infusion significantly elevated blood pressure compared with vehicle-infused rats (n = 6). Treatment with temocapril or olmesartan suppressed Ang I-induced hypertension. Temocapril suppressed both plasma and renal ACE activity. Ang I infusion increased Ang II content in the kidney. Interestingly, temocapril failed to reduce the level of Ang II in the kidney, while olmesartan markedly suppressed an increase in renal Ang II levels. These results suggest a limitation of temocapril and a benefit of olmesartan to inhibit the renal renin-angiotensin system and suggest the possible existence of an ACE inhibitor-insensitive pathway that increases Ang II levels in rat kidney.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II/urine , Angiotensin I/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Kidney/metabolism , Tetrazoles/pharmacology , Thiazepines/pharmacology , Angiotensin I/administration & dosage , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Hypertension/etiology , Imidazoles/therapeutic use , Male , Peptidyl-Dipeptidase A/metabolism , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Tetrazoles/therapeutic use , Thiazepines/therapeutic use
12.
J Pharmacol Sci ; 121(2): 95-102, 2013.
Article in English | MEDLINE | ID: mdl-23337436

ABSTRACT

We examined the effects of angiotensin II AT1-receptor blockade with olmesartan on high fat (HF) diet-induced vascular oxidative stress and endothelial dysfunction in normal salt (NS) diet-fed Dahl salt-sensitive (DSS) rats. Treatment with NS + HF diet (32% crude fat, 0.3% NaCl) for 20 weeks significantly increased blood pressure in DSS rats. NS + HF diet-fed DSS rats also showed higher plasma levels of thiobarbituric acid-reactive substances, aortic superoxide production, and mRNA levels of p22(phox) and gp91(phox) in aortic tissues than NS diet-fed DSS rats. Furthermore, acetylcholine-induced vasorelaxation of aorta from NS + HF diet-fed DSS rats was significantly reduced. In NS + HF diet-fed DSS rats, treatment with olmesartan medoxomil (10 mg/kg per day, p.o.) and hydralazine (25 mg/kg per day, p.o.) similarly decreased blood pressure. However, in these animals, only olmesartan normalized plasma levels of thiobarbituric acid-reactive substances, vascular superoxide in aortic tissues, and acetylcholine-induced vasorelaxation. These data indicate that HF diet-induced hypertension is associated with vascular oxidative stress and endothelial dysfunction in NS diet-treated DSS rats. Inhibition of angiotensin II AT1 receptors may elicit beneficial effects on HF-induced hypertension and vascular injury in subjects that have genetically enhanced sodium-sensitive blood pressure.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Diet, High-Fat/adverse effects , Endothelium, Vascular/drug effects , Oxidative Stress/drug effects , Receptor, Angiotensin, Type 1/metabolism , Animals , Antihypertensive Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Endothelium, Vascular/metabolism , Hydralazine/pharmacology , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Imidazoles/pharmacology , Male , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , NADPH Oxidases/metabolism , Olmesartan Medoxomil , Rats , Rats, Inbred Dahl , Superoxides/metabolism , Tetrazoles/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism , Vasodilation/drug effects
13.
J Hypertens ; 29(11): 2210-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21934531

ABSTRACT

BACKGROUND: Neurovascular protection against cerebral ischemia is not consistently observed with a postischemia hypotensive dose of candesartan. The aim of this study was to determine the levels of brain angiotensin II after reperfusion and the efficacy and therapeutic time window of postischemic treatments with hypotensive doses of candesartan for the treatment of cerebral ischemia. METHOD: Occlusions of the right middle cerebral artery (60 min) followed by reperfusion were performed using the thread method under halothane anesthesia in Sprague-Dawley (SD) rats. Protein levels of brain angiotensin II and mRNA levels of renin-angiotensin system components were evaluated following reperfusion (n=184 in total). Low-dose or high-dose treatments with candesartan cilexetil (1 or 10 mg/kg per day, respectively) were administered orally immediately following reperfusion once daily for 4 or 7 days (n = 119 in total). An additional group was treated with low-dose candesartan cilexetil after a 12-h delay based on the brain angiotensin II levels (n = 14). RESULTS: Levels of brain angiotensin II transiently increased 4-12 h after reperfusion, which followed an increase in angiotensinogen mRNA. Candesartan cilexetil treatments significantly reduced blood pressure (BP) in rats administered the high dose and moderately in rats receiving the low dose. A low dose of candesartan cilexetil reduced the infarct size, cerebral edema, and neurological deficits, whereas the high-dose treatments showed limited reductions. Furthermore, oxidative stress following reperfusion was reduced with the low-dose treatments. The therapeutic time window was open for at least 12 h after reperfusion when brain angiotensin II levels had peaked. CONCLUSION: Postischemic treatments using low hypotensive doses of candesartan cilexetil provided protection against cerebral ischemic injury and may have a clinically relevant therapeutic time window.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Ischemia/pathology , Tetrazoles/pharmacology , Angiotensin II/blood , Angiotensinogen/metabolism , Animals , Biphenyl Compounds/pharmacology , Blood Pressure , Brain/metabolism , Brain/pathology , Brain Edema/pathology , Hypotension , Rats , Rats, Sprague-Dawley , Time Factors
14.
Hypertens Res ; 34(4): 479-83, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270817

ABSTRACT

Angiotensin II (Ang II)-induced astrocyte senescence may be involved in cerebral ischemic injury and age-associated neurodegenerative disease. This study was conducted to determine the roles of reactive oxygen species production in Ang II-induced cellular senescence in cultured human astrocytes. Human astrocytes were stimulated with Ang II either with or without an angiotensin type 1 receptor blocker, CV11974, or an antioxidant, tempol. Application of Ang II to human astrocytes resulted in a concentration-dependent increase in staining for dihydroethidium. Ang II (100 nM for 30 min) increased the translocation of two cytosolic components of NADPH oxidase, p47phox and p67phox, to the cell membrane and formation of the complex of p47phox, p67phox and p22phox. Ang II concentration-dependently induced an increase in ß-galactosidase staining. Pretreatment with CV11974 (100 nM) or tempol (3 mM) abolished Ang II-induced astrocyte ß-galactosidase staining. Moreover, Ang II significantly upregulated p16 mRNA expression, which was inhibited by pretreatment with CV11974 or tempol. These findings indicate that superoxide production contributes to Ang II-induced astrocyte senescence.


Subject(s)
Angiotensin II/pharmacology , Astrocytes/cytology , Astrocytes/metabolism , Cellular Senescence/drug effects , Reactive Oxygen Species/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Antioxidants/pharmacology , Astrocytes/drug effects , Benzimidazoles/pharmacology , Biphenyl Compounds , Cells, Cultured , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Drug , Humans , NADPH Oxidases/metabolism , Spin Labels , Superoxides/metabolism , Tetrazoles/pharmacology
15.
Am J Hypertens ; 24(3): 362-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21164491

ABSTRACT

BACKGROUND: The present study tested the hypothesis that inappropriate activation of the brain renin-angiotensin system (RAS) contributes to the pathogenesis of blood-brain barrier (BBB) disruption and cognitive impairment during development of salt-dependent hypertension. Effects of an angiotensin II (AngII) type-1 receptor blocker (ARB), at a dose that did not reduce blood pressure, were also examined. METHODS: Dahl salt-sensitive (DSS) rats at 6 weeks of age were assigned to three groups: low-salt diet (DSS/L; 0.3% NaCl), high-salt diet (DSS/H; 8% NaCl), and high-salt diet treated with ARB, olmesartan at 1 mg/kg. RESULTS: DSS/H rats exhibited hypertension, leakage from brain microvessels in the hippocampus, and impaired cognitive functions, which were associated with increased brain AngII levels, as well as decreased mRNA levels of tight junctions (TJs) and collagen-IV in the hippocampus. In DSS/H rats, olmesartan treatment, at a dose that did not alter blood pressure, restored the cognitive decline, and ameliorated leakage from brain microvessels. Olmesartan also decreased brain AngII levels and restored mRNA expression of TJs and collagen-IV in DSS/H rats. CONCLUSIONS: These results suggest that during development of salt-dependent hypertension, activation of the brain RAS contributes to BBB disruption and cognitive impairment. Treatment with an ARB could elicit neuroprotective effects in cognitive disorders by preventing BBB permeability, which is independent of blood pressure changes.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Blood-Brain Barrier/drug effects , Cognition/drug effects , Hypertension/drug therapy , Angiotensin II/analysis , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Body Weight/drug effects , Capillary Permeability/drug effects , Corpus Callosum/metabolism , Hippocampus/metabolism , Hypertension/metabolism , Hypertension/psychology , Imidazoles/pharmacology , Imidazoles/therapeutic use , Immunohistochemistry , Male , Platelet Endothelial Cell Adhesion Molecule-1/analysis , RNA, Messenger/analysis , Rats , Rats, Inbred Dahl , Systole/drug effects , Tetrazoles/pharmacology , Tetrazoles/therapeutic use
16.
Hypertens Res ; 33(12): 1250-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20927110

ABSTRACT

Angiotensin II (AngII) stimulates vascular smooth muscle cell (VSMC) proliferation; however, the effect of AngII on cell proliferation in the presence of mechanical force is not clear. We investigated the mechanism of AngII-induced cell proliferation mediated by mechanical stretch in VSMCs of both normotensive and hypertensive rats. VSMCs obtained from the thoracic aortas of 8-week-old Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHR) were stretched by a Flex culture system. Mechanical stretch significantly upregulated protein expression of AngII type 1 (AT1) receptor, epidermal growth factor (EGF) receptor and mitogen-activated protein kinase phosphatase-1 in both SHR and WKY VSMCs; however, there was no significant difference in these changes between the cells from SHR and WKY. Mechanical stretch attenuated AngII-induced phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, ERK kinase (MEK) and EGF receptor; it also attenuated [³H] thymidine incorporation and cell proliferation in VSMC of WKY. In contrast, the effects of AngII were augmented by mechanical stretch in VSMC of SHR. AngII-induced ERK 1/2 phosphorylation and cell proliferation in SHR were inhibited by pretreatment with an AT1 receptor blocker, candesartan and an inhibitor of MEK, PD98059. Moreover, pretreatment with an EGF receptor tyrosine kinase inhibitor, AG1478, also blocked upregulation of AngII-induced ERK 1/2 phosphorylation induced by stretch in SHR VSMCs. This study demonstrates that mechanical stretch augments SHR VSMC proliferation through an AT1/EGF receptor/ERK-dependent pathway. These findings may provide new insights into the signaling mechanisms whereby AngII exerts its growth-promoting effects on vasculature in a hypertensive state.


Subject(s)
Angiotensin II/pharmacology , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Stress, Mechanical , Analysis of Variance , Animals , Blotting, Western , Cell Count , Cells, Cultured , ErbB Receptors/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
17.
J Hypertens ; 28(11): 2289-98, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20706133

ABSTRACT

OBJECTIVE: Clinical studies indicate that the remission, regression or both of nephrotic-range albuminuria are exerted by angiotensin II receptor blockers (ARBs) in diabetes. The current study was performed to test the hypothesis that these effects of ARBs are associated with regression of glomerular podocyte injury. METHODS: We examined the effects of an ARB, olmesartan, on glomerular podocyte injury in type 2 diabetic Otsuka-Long-Evans-Tokushima-Fatty rats with overt albuminuria. RESULTS: At baseline (55-week-old), diabetic Otsuka-Long-Evans-Tokushima-Fatty rats showed severe albuminuria with desmin-positive areas (an index of podocyte injury) in both superficial and juxtamedullary glomeruli, and podocyte injury was much greater in juxtamedullary than in superficial glomeruli. At 75-week-old, Otsuka-Long-Evans-Tokushima-Fatty rats had developed more severe albuminuria and superficial glomerular podocyte injury, whereas juxtamedullary glomerular podocyte injury did not advance further. Olmesartan (10 mg/kg per day) decreased albuminuria and superficial glomerular desmin staining to levels that were lower than those at baseline, whereas advanced juxtamedullary glomerular podocyte injury was not changed. CONCLUSION: The current study demonstrates for the first time that juxtamedullary glomerular podocyte injury reaches a severe condition at an earlier time than superficial glomerular podocyte injury during the progression of overt albuminuria in type 2 diabetic rats. Our data also support the hypothesis that the antialbuminuric effects of ARBs are associated with regression of superficial glomerular podocyte injury in type 2 diabetes with nephrotic-range albuminuria.


Subject(s)
Albuminuria/pathology , Angiotensin II/metabolism , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 2/therapy , Animals , Disease Models, Animal , Kidney/pathology , Kidney Glomerulus/pathology , Male , Podocytes/pathology , Rats , Rats, Long-Evans , Remission Induction , Sclerosis/pathology , Treatment Outcome
18.
Hypertens Res ; 33(2): 161-4, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19942928

ABSTRACT

The renin-angiotensin system has an important function in the regulation of blood pressure as well as in pathophysiological processes in the central nervous system. We examined the effects of the angiotensin receptor blocker candesartan (10 mg kg(-1) per day, p.o.) on brain angiotensin II levels in angiotensin II-infused hypertensive rats. Angiotensin II or vehicle was infused subcutaneously for 14 days in Sprague-Dawley rats. Angiotensin II infusion resulted in increased blood pressure, an effect that was blocked by candesartan treatment. There was no effect of the angiotensin II infusion on Angiotensin II levels in the brain or on blood-brain barrier permeability. Brain tissue angiotensinogen and angiotensin converting enzyme mRNA levels were not changed by angiotensin II infusion but were decreased by candesartan treatment. At 2 weeks of treatment, CV11974, an active form of candesartan, was detectable in the plasma but was not detectable in brain tissue. These data suggest that treatment with candesartan decreases brain angiotensin II by decreasing brain angiotensinogen and angiotensin converting enzyme gene expression.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II/analysis , Benzimidazoles/pharmacology , Brain Chemistry/drug effects , Renin-Angiotensin System/drug effects , Tetrazoles/pharmacology , Angiotensin II/blood , Animals , Benzimidazoles/metabolism , Biphenyl Compounds , Blood Pressure/drug effects , Blood-Brain Barrier , Body Weight/drug effects , Down-Regulation , Male , Permeability , Rats , Rats, Sprague-Dawley , Tetrazoles/metabolism
19.
Exp Physiol ; 94(9): 1016-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19502292

ABSTRACT

Recent studies have shown that blocking non-proteolytically activated prorenin with a decoy peptide for the handle region of the prorenin prosegment (HRP) inhibits the development of microvascular complications in diabetic animals. In the present study, we investigated whether non-proteolytic activation of prorenin contributes to the development of fructose-induced insulin resistance. Rats were fed a standard diet (n = 10), a 60% high fructose diet (n = 16), or a high fructose diet + HRP (0.1 mg kg(-1) day(-1), n = 16) for 10 weeks. Fructose-fed rats showed higher systolic blood pressure (SBP), fasting plasma triglycerides, total cholesterol and insulin levels; which, except for SBP, were suppressed by HRP. The responses of plasma glucose and insulin levels to oral glucose loading were significantly greater in fructose-fed rats than in standard diet-fed rats. The HRP normalized the enhanced responses of plasma glucose and insulin levels that were observed in fructose-fed rats. Moreover, HRP suppressed the enhanced prorenin activation and angiotensin II formation in the soleus muscle of fructose-fed rats. These data suggest that local angiotensin II generation in skeletal muscle, induced by non-proteolytic activation of prorenin, contributes to the development of insulin resistance induced by a high fructose diet.


Subject(s)
Dietary Carbohydrates/administration & dosage , Fructose/administration & dosage , Insulin Resistance/physiology , Renin/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/genetics , Angiotensin II/biosynthesis , Animals , Base Sequence , Blood Glucose/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Cell Size/drug effects , Cholesterol/blood , Dietary Carbohydrates/adverse effects , Enzyme Activation/drug effects , Fatty Acids, Nonesterified/blood , Fructose/adverse effects , Insulin/blood , Insulin Resistance/genetics , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Oligopeptides/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Triglycerides/blood , Prorenin Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...