Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Lasers Med Sci ; 39(1): 122, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38703271

ABSTRACT

Pulsed dye lasers are used effectively in the treatment of psoriasis with long remission time and limited side effects. It is, however, not completely understood which biological processes underlie its favorable outcome. Pulsed dye laser treatment at 585-595 nm targets hemoglobin in the blood, inducing local hyperthermia in surrounding blood vessels and adjacent tissues. While the impact of destructive temperatures on blood vessels has been well studied, the effects of lower temperatures on the function of several cell types within the blood vessel wall and its periphery are not known. The aim of our study is to assess the functionality of isolated blood vessels after exposure to moderate hyperthermia (45 to 60°C) by evaluating the function of endothelial cells, smooth muscle cells, and vascular nerves. We measured blood vessel functionality of rat mesenteric arteries (n=19) by measuring vascular contraction and relaxation before and after heating vessels in a wire myograph. To this end, we elicited vascular contraction by addition of either high potassium solution or the thromboxane analogue U46619 to stimulate smooth muscle cells, and electrical field stimulation (EFS) to stimulate nerves. For measurement of endothelium-dependent relaxation, we used methacholine. Each vessel was exposed to one temperature in the range of 45-60°C for 30 seconds and a relative change in functional response after hyperthermia was determined by comparison with the response per stimulus before heating. Non-linear regression was used to fit our dataset to obtain the temperature needed to reduce blood vessel function by 50% (Half maximal effective temperature, ET50). Our findings demonstrate a substantial decrease in relative functional response for all three cell types following exposure to 55°C-60°C. There was no significant difference between the ET50 values of the different cell types, which was between 55.9°C and 56.9°C (P>0.05). Our data show that blood vessel functionality decreases significantly when exposed to temperatures between 55°C-60°C for 30 seconds. The results show functionality of endothelial cells, smooth muscle cells, and vascular nerves is similarly impaired. These results help to understand the biological effects of hyperthermia and may aid in tailoring laser and light strategies for selective photothermolysis that contribute to disease modification of psoriasis after pulsed dye laser treatment.


Subject(s)
Lasers, Dye , Animals , Rats , Male , Lasers, Dye/therapeutic use , Myocytes, Smooth Muscle/physiology , Myocytes, Smooth Muscle/radiation effects , Vasodilation/radiation effects , Vasodilation/physiology , Temperature , Muscle, Smooth, Vascular/radiation effects , Muscle, Smooth, Vascular/physiology , Endothelial Cells/radiation effects , Endothelial Cells/physiology , Vasoconstriction/radiation effects , Vasoconstriction/physiology , Endothelium, Vascular/radiation effects , Rats, Wistar
2.
PLoS One ; 16(10): e0257896, 2021.
Article in English | MEDLINE | ID: mdl-34610026

ABSTRACT

INTRODUCTION: Peripheral artery disease (PAD) is a highly morbid condition in which impaired blood flow to the limbs leads to pain and tissue loss. Previously we identified 670 nm electromagnetic energy (R/NIR) to increase nitric oxide levels in cells and tissue. NO elicits relaxation of smooth muscle (SMC) by stimulating potassium efflux and membrane hyperpolarization. The actions of energy on ion channel activity have yet to be explored. Here we hypothesized R/NIR stimulates vasodilation through activation of potassium channels in SMC. METHODS: Femoral arteries or facial arteries from C57Bl/6 and Slo1-/- mice were isolated, pressurized to 60 mmHg, pre-constricted with U46619, and irradiated twice with energy R/NIR (10 mW/cm2 for 5 min) with a 10 min dark period between irradiations. Single-channel K+ currents were recorded at room temperature from cell-attached and excised inside-out membrane patches of freshly isolated mouse femoral arterial muscle cells using the patch-clamp technique. RESULTS: R/NIR stimulated vasodilation requires functional activation of the large conductance potassium channels. There is a voltage dependent outward current in SMC with light stimulation, which is due to increases in the open state probability of channel opening. R/NIR modulation of channel opening is eliminated pharmacologically (paxilline) and genetically (BKca α subunit knockout). There is no direct action of light to modulate channel activity as excised patches did not increase the open state probability of channel opening. CONCLUSION: R/NIR vasodilation requires indirect activation of the BKca channel.


Subject(s)
Electromagnetic Radiation , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/radiation effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/radiation effects , Signal Transduction/radiation effects , Vasodilation/radiation effects , Animals , Electric Stimulation/methods , Electric Stimulation Therapy/methods , Femoral Artery/metabolism , Gene Knockout Techniques , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Membrane Potentials/radiation effects , Mice , Mice, Knockout , Nitric Oxide/metabolism , Patch-Clamp Techniques , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/therapy
3.
J Am Heart Assoc ; 10(14): e020712, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34227406

ABSTRACT

Background Chronic inflammation through cellular senescence, known as the senescence-associated secretory phenotype, is a mechanism of various organ diseases, including atherosclerosis. Particularly, ionizing radiation (IR) contributes to cellular senescence by causing DNA damage. Although previous clinical studies have demonstrated that radiotherapy causes atherosclerosis as a long-term side effect, the detailed mechanism is unclear. This study was conducted to investigate the relationship between radiation-induced atherosclerosis and senescence-associated secretory phenotype in murine carotid arteries. Methods and Results Partial ligation of the left carotid artery branches in 9-week-old male apolipoprotein E-deficient mice was performed to induce atherosclerosis. The mice received total body irradiation at a dose of 6 Gy using gamma rays at 2 weeks post operation. We compared the samples collected 4 weeks after IR with unirradiated control samples. The IR and control groups presented pathologically progressive lesions in 90.9% and 72.3% of mice, respectively. Plaque volume, macrophage accumulation, and phenotype switching of vascular smooth muscle cells were advanced in the IR group. Irradiated samples showed increased persistent DNA damage response (53BP1 [p53 binding protein 1]), upregulated cyclin-dependent kinase inhibitors (p16INK4a and p21), and elevated inflammatory chemokines expression (monocyte chemotactic protein-1, keratinocyte-derived chemokine, and macrophage inflammatory protein 2). Conclusions IR promoted plaque growth in murine carotid arteries. Our findings support the possibility that senescence-associated secretory phenotype aggravates atherogenesis in irradiated artery. This mice model might contribute to mechanism elucidation of radiation-induced atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Carotid Artery, Common/radiation effects , Macrophages/pathology , Myocytes, Smooth Muscle/radiation effects , Plaque, Atherosclerotic/etiology , Radiation Injuries, Experimental/complications , Regional Blood Flow/physiology , Animals , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Carotid Artery, Common/pathology , Cellular Senescence/radiation effects , Chemokines/biosynthesis , Disease Progression , Dose-Response Relationship, Radiation , Macrophages/metabolism , Macrophages/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology
4.
J Allergy Clin Immunol Pract ; 7(1): 71-80, 2019 01.
Article in English | MEDLINE | ID: mdl-30193939

ABSTRACT

Bronchial thermoplasty (BT) delivers targeted radiofrequency energy to bronchial airway walls and results in the partial ablation of the airway smooth muscle that is responsible for bronchoconstriction. It is approved for the treatment of severe persistent asthma. Multiple, large clinical trials including a recent "real-world" study demonstrate significant improvements in asthma-related quality of life, reduction in asthma exacerbations, emergency department visits, and hospitalizations after BT that is sustained out to 5 years. In this article, we review the state of the art of BT treatment in severe persistent asthma and share a decade of BT research and clinical experience. We share our personal experience and introduce the three "I"s (identification, implementation, and intense follow-up) that we believe promote successful patient outcomes and help build a successful BT program.


Subject(s)
Asthma/therapy , Bronchi/pathology , Bronchial Thermoplasty/methods , Myocytes, Smooth Muscle/radiation effects , Radiofrequency Therapy/methods , Bronchoconstriction , Clinical Trials as Topic , Disease Progression , Humans , Myocytes, Smooth Muscle/physiology , Quality of Life , Radiofrequency Ablation , Treatment Outcome
5.
JCI Insight ; 3(15)2018 08 09.
Article in English | MEDLINE | ID: mdl-30089722

ABSTRACT

The long-term adverse effects of radiotherapy on cardiovascular disease are well documented. However, the underlying mechanisms responsible for this increased risk are poorly understood. Previous studies using rigorous smooth muscle cell (SMC) lineage tracing have shown abundant SMC investment into atherosclerotic lesions, where SMCs contribute to the formation of a protective fibrous cap. Studies herein tested whether radiation impairs protective adaptive SMC responses during vascular disease. To do this, we exposed SMC lineage tracing (Myh11-ERT2Cre YFP+) mice to lethal radiation (1,200 cGy) followed by bone marrow transplantation prior to atherosclerosis development or vessel injury. Surprisingly, following irradiation, we observed a complete loss of SMC investment in 100% of brachiocephalic artery (BCA), carotid artery, and aortic arch lesions. Importantly, this was associated with a decrease in multiple indices of atherosclerotic lesion stability within the BCA. Interestingly, we observed anatomic heterogeneity, as SMCs accumulated normally into lesions of the aortic root and abdominal aorta, suggesting that SMC sensitivity to lethal irradiation occurs in blood vessels of neural crest origin. Taken together, these results reveal an undefined and unintended variable in previous studies using lethal irradiation and may help explain why patients exposed to radiation have increased risk for cardiovascular disease.


Subject(s)
Atherosclerosis/pathology , Brachiocephalic Trunk/radiation effects , Muscle, Smooth, Vascular/radiation effects , Myocytes, Smooth Muscle/radiation effects , Animals , Aorta, Abdominal/pathology , Aorta, Abdominal/radiation effects , Atherosclerosis/etiology , Bone Marrow/radiation effects , Bone Marrow Transplantation , Brachiocephalic Trunk/pathology , Cell Differentiation/radiation effects , Disease Models, Animal , Humans , Male , Mice , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/cytology , Whole-Body Irradiation
6.
J Vasc Access ; 19(2): 153-161, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29192723

ABSTRACT

PURPOSE: To study the effect of x-ray radiotherapy on vascular smooth muscle cells (VSMCs) and elucidate the mechanisms in preventing neointimal hyperplasia of prosthetic vascular grafts. MATERIALS AND METHODS: In model I, twelve mongrel dogs underwent revascularization with prosthetic grafts and half the dogs underwent irradiation of the grafts at 28 Gy. In model II, human VSMCs (hVSMCs) were maintained and divided into six groups to which external radiation was applied at six different doses: 0 Gy, 2 Gy, 8 Gy, 16 Gy, 24 Gy and 30 Gy. In both models, specimens were harvested and examined by using morphological, immunological, cellular and molecular methods. RESULTS: After irradiation, the neointima thickness was significantly lower in irradiated groups (p≤0.01). The radiotherapy could up-regulate p27kip1, and down-regulate proliferating cell nuclear antigen (PCNA) and S phase kinase associated protein 2 (Skp2). X-ray irradiation inhibits the proliferation of hVSMCs via acting on G1/S phase of cell cycle. The apoptosis of hVSMCs increased significantly with dose and time. The expression of PCNA and Skp2 were decreased after a first increasing trend with dose, but had a significant negative correlation with time. The expression of p27kip1 had a significant positive correlation with dose and time. CONCLUSIONS: Postoperative external fractionated irradiation after prosthetic vessel replacement of the abdominal aorta suppressed the development of hyperplasia in the graft neointima in the short term. There was a prominent time- and dose-dependent inhibition of VSMC proliferation by radiation when it was administered.


Subject(s)
Aorta, Abdominal/radiation effects , Aorta, Abdominal/surgery , Apoptosis/drug effects , Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Cell Proliferation/radiation effects , Muscle, Smooth, Vascular/radiation effects , Muscle, Smooth, Vascular/surgery , Myocytes, Smooth Muscle/radiation effects , Neointima , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Blood Vessel Prosthesis Implantation/adverse effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dogs , Dose-Response Relationship, Radiation , Humans , Hyperplasia , Models, Animal , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Proliferating Cell Nuclear Antigen/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Time Factors
7.
Lasers Med Sci ; 32(9): 2121-2127, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28983687

ABSTRACT

Vascular restenosis after injury of blood vessel has been implicated in various responses including apoptosis, migration, and proliferation in vascular smooth muscle cells (VSMCs) stimulated by diverse growth factors underlying platelet-derived growth factor (PDGF). Previous studies evaluated the effects of low-power laser (LPL) irradiation over various wavelength ranges on VSMC events in normal and pathologic states. However, whether VSMC responses are affected by LPL irradiation remains unclear. The purpose of this study is to explore the effects of LPL (green diode laser 532-nm pulsed wave of 300 mW at a spot diameter of 1 mm) irradiation on the responses, apoptosis, migration, and proliferation of VSMCs. The effect of LPL irradiation was tested on VSMCs through cytotoxicity, proliferation, migration, and apoptotic assays. Aortic ring assay was used to assess the effect of LPL irradiation on aortic sprout outgrowth. Protein expression levels were determined by western blotting. LPL irradiation did not affect VSMC viability but slightly attenuated PDGF-BB-induced proliferation in VSMCs. In addition, LPL irradiation inhibited PDGF-BB-evoked migration of VSMCs. Aortic sprout outgrowth in response to PDGF-BB was diminished in cells treated with LPL. In contrast, LPL irradiation evoked apoptosis in VSMCs in the presence of PDGF-BB. Similarly, activation of caspase-3 and Bax, as well as p38 mitogen-activated protein kinase (MAPK), in VSMCs treated with PDGF-BB was enhanced by exposure to LPL. These findings indicate that LPL irradiation induces vascular apoptosis via p38 MAPK activation and simultaneously inhibits VSMC proliferation and migration in response to PDGF-BB.


Subject(s)
Apoptosis/drug effects , Cell Movement/drug effects , Low-Level Light Therapy , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/radiation effects , Proto-Oncogene Proteins c-sis/pharmacology , Animals , Aorta/cytology , Becaplermin , Cell Proliferation/drug effects , Cells, Cultured , Male , Myocytes, Smooth Muscle/drug effects , Rats, Sprague-Dawley
8.
Lasers Med Sci ; 32(8): 1737-1746, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28653257

ABSTRACT

Skin flap grafting is a form of transplantation widely used in plastic surgery. However, ischemia/reperfusion injury is the main factor which reduces the survival rate of flaps following grafting. We investigated whether photobiomodulation (PBM) precondition prior to human adipose-derived stromal cell (hASC) spheroid (PBM-spheroid) transplantation improved skin tissue functional recovery by the stimulation of angiogenesis and tissue regeneration in skin flap of mice. The LED had an emission wavelength peaked at 660 ± 20 nm (6 J/cm2, 10 mW/cm2). The expression of angiogenic growth factors in PBM-spheroid hASCs was much greater than that of not-PBM-treated spheroid or monolayer-cultured hASCs. From immunochemical staining analysis, the hASCs of PBM-spheroid were CD31+, KDR+, and CD34+, whereas monolayer-cultured hASCs were negative for these markers. To evaluate the therapeutic effect of hASC PBM-spheroid in vivo, PBS, monolayer-cultured hASCs, and not-PBM-spheroid were transplanted into a skin flap model. The animals were observed for 14 days. The PBM-spheroid hASCs transplanted into the skin flap ischemia differentiated into endothelial cells and remained differentiated. Transplantation of PBM-spheroid hASCs into the skin flap ischemia significantly elevated the density of vascular formations through angiogenic factors released by the skin flap ischemia and enhanced tissue regeneration at the lesion site. Consistent with these results, the transplantation of PBM-spheroid hASCs significantly improved functional recovery compared with PBS, monolayer-cultured hASCs, and not-PBM-spheroid treatment. These findings suggest that transplantation of PBM-spheroid hASCs may be an effective stem cell therapy for the treatment of skin flap ischemia.


Subject(s)
Adipose Tissue/cytology , Ischemia/therapy , Low-Level Light Therapy , Regeneration/radiation effects , Skin/blood supply , Spheroids, Cellular/cytology , Stem Cells/cytology , Surgical Flaps/blood supply , Animals , Cell Differentiation/radiation effects , Cell Survival/radiation effects , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/radiation effects , Epithelial Cells/cytology , Epithelial Cells/radiation effects , Humans , Ischemia/pathology , Mice , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/radiation effects , Neovascularization, Physiologic/radiation effects , Skin/pathology , Spheroids, Cellular/radiation effects , Stem Cell Transplantation , Stem Cells/radiation effects , Wound Healing
9.
Acta Biomater ; 52: 171-186, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27884774

ABSTRACT

Arresting or regressing growth of abdominal aortic aneurysms (AAAs), localized expansions of the abdominal aorta are contingent on inhibiting chronically overexpressed matrix metalloproteases (MMPs)-2 and -9 that disrupt elastic matrix within the aortic wall, concurrent with providing a stimulus to augmenting inherently poor auto-regeneration of these matrix structures. In a recent study we demonstrated that localized, controlled and sustained delivery of doxycycline (DOX; a tetracycline-based antibiotic) from poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs), enhances elastic matrix deposition and MMP-inhibition at a fraction of the therapeutically effective oral dose. The surface functionalization of these NPs with cationic amphiphiles, which enhances their arterial uptake, was also shown to have pro-matrix regenerative and anti-MMP effects independent of the DOX. Based on the hypothesis that the incorporation of superparamagnetic iron oxide NPs (SPIONs) within these PLGA NPs would enhance their targetability to the AAA site under an applied external magnetic field, we sought to evaluate the functional effects of NPs co-encapsulating DOX and SPIONs (DOX-SPION NPs) on elastic matrix regeneration and MMP synthesis/activity in vitro within aneurysmal smooth muscle cell (EaRASMC) cultures. The DOX-SPION NPs were mobile under an applied external magnetic field, while enhancing elastic matrix deposition 1.5-2-fold and significantly inhibiting MMP-2 synthesis and MMP-2 and -9 activities, compared to NP-untreated control cultures. These results illustrate that the multifunctional benefits of NPs are maintained following SPION co-incorporation. Additionally, preliminary studies carried out demonstrated enhanced targetability of SPION-loaded NPs within proteolytically-disrupted porcine carotid arteries ex vivo, under the influence of an applied external magnetic field. Thus, this dual-agent loaded NP system proffers a potential non-surgical option for treating small growing AAAs, via controlled and sustained drug release from multifunctional, targetable nanocarriers. STATEMENT OF SIGNIFICANCE: Proactive screening of high risk elderly patients now enables early detection of abdominal aortic aneurysms (AAAs). There are no established drug-based therapeutic alternatives to surgery for AAAs, which is unsuitable for many elderly patients, and none which can achieve restore disrupted and lost elastic matrix in the AAA wall, which is essential to achieve growth arrest or regression. We have developed a first generation design of polymer nanoparticles (NPs) for AAA tissue localized delivery of doxycycline, a modified tetracycline drug at low micromolar doses at which it provides both pro-elastogenic and anti-proteolytic benefits that can augment elastic matrix regenerative repair. The nanocarriers themselves are also uniquely chemically functionalized on their surface to also provide them pro-elastin-regenerative & anti-matrix degradative properties. To provide an active driving force for efficient uptake of intra-lumenally infused NPs to the AAA wall, in this work, we have rendered our polymer NPs mobile in an applied magnetic field via co-incorporation of super-paramagnetic iron oxide NPs. We demonstrate that such modifications significantly improve wall uptake of the NPs with no significant changes to their physical properties and regenerative benefits. Such NPs can potentially stimulate structural repair in the AAA wall following one time infusion to delay or prevent AAA growth to rupture. The therapy can provide a non-surgical treatment option for high risk AAA patients.


Subject(s)
Delayed-Action Preparations/administration & dosage , Dextrans/administration & dosage , Doxycycline/administration & dosage , Magnetite Nanoparticles/administration & dosage , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/radiation effects , Regeneration/drug effects , Animals , Cells, Cultured , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/radiation effects , Dextrans/radiation effects , Doxycycline/chemistry , Extracellular Matrix/drug effects , Extracellular Matrix/radiation effects , Magnetic Fields , Magnetite Nanoparticles/radiation effects , Male , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Nanocapsules/radiation effects , Radiation Dosage , Rats , Rats, Sprague-Dawley , Regeneration/radiation effects
10.
Drug Discov Ther ; 10(2): 79-81, 2016.
Article in English | MEDLINE | ID: mdl-27087553

ABSTRACT

Whole brain radiation therapy for the treatment of tumors can sometimes cause cognitive impairment. Memory deficits were noted in up to 50% of treated patients over a short period of several months. In addition, an increased rate of dementia in young patients has been noted over the longer term, i.e. years. A deficit in neurogenesis after irradiation has been postulated to be the main cause of cognitive decline in patients, but recent data on irradiation therapy for limited parts of the brain appear to indicate other possibilities. Irradiation can directly damage various types of cells other than neuronal stem cells. However, this paper will focus on injury to brain vasculature leading to cognitive decline since vessels represent a better therapeutic target for drug development than other cells in the brain because of the blood-brain barrier.


Subject(s)
Blood Vessels/injuries , Blood Vessels/radiation effects , Brain/radiation effects , Cognition Disorders/etiology , Neoplasms/radiotherapy , Radiotherapy/adverse effects , Blood Vessels/pathology , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/radiation effects
11.
Sci Rep ; 6: 21683, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26899550

ABSTRACT

Paclitaxel (PTX) has been recognized as a promising drug for intervention of vascular reconstructions. However, it is still difficult to achieve local drug delivery in a spatio-temporally controllable manner under real-time image guidance. Here, we introduce an ultrasound (US) triggered image-guided drug delivery approach to inhibit vascular reconstruction via paclitaxel (PTX)-loaded microbubbles (PLM) in a rabbit iliac balloon injury model. PLM was prepared through encapsulating PTX in the shell of lipid microbubbles via film hydration and mechanical vibration technique. Our results showed PLM could effectively deliver PTX when exposed to US irradiation and result in significantly lower viability of vascular smooth muscle cells. Ultrasonographic examinations revealed the US signals from PLM in the iliac artery were greatly increased after intravenous administration of PLM, making it possible to identify the restenosis regions of iliac artery. The in vivo anti-restenosis experiments with PLM and US greatly inhibited neointimal hyperplasia at the injured site, showing an increased lumen area and reduced the ratio of intima area and the media area (I/M ratio). No obvious functional damages to liver and kidney were observed for those animals. Our study provided a promising approach to realize US triggered image-guided PTX delivery for therapeutic applications against iliac restenosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Constriction, Pathologic/therapy , Drug Delivery Systems/methods , Iliac Artery/drug effects , Microbubbles , Paclitaxel/pharmacology , Animals , Constriction, Pathologic/pathology , Drug Compounding , Drug Delivery Systems/instrumentation , Iliac Artery/pathology , Muscle, Smooth, Vascular/radiation effects , Myocytes, Smooth Muscle/radiation effects , Rabbits , Ultrasonic Waves
12.
Photomed Laser Surg ; 34(11): 509-515, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26594838

ABSTRACT

OBJECTIVE: The aim of the study was to investigate the differentiation potential of adipose-derived stem cells (ADSCs) when cocultured with smooth muscle cells (SMCs), and to determine the role of low-intensity laser irradiation (LILI). BACKGROUND DATA: ADSCs isolated from adipose tissue are isolated with ease and in large amounts. SMCs constitute most parts of the intestinal, urinary, reproductive, and cardiovascular systems. LILI has been found to have positive effects on different cell types, including ADSCs. METHODS: The study used ADSCs (Stempro Adipose Derived Stem Cells-R7788-115) and SMCs (SKU-T-1 American Type Culture Collection HTB-114) cell lines. These cell lines were cocultured in a 1:1 ratio with and without growth factors and then exposed to LILI using 636 nm at 5 J/cm2. RESULTS: Cell viability and proliferation increased significantly in the cocultured groups that were exposed to LILI alone, as well as in combination with growth factors. Further, there was a significant decrease in the expression of stem cell markers with a concomitant increase in SMC markers. CONCLUSIONS: These results suggest that ADSCs have the ability to differentiate into SMCs when cocultured with SMCs, whereas LILI potentially augments the differentiation potential and need. This further highlights the significant role that LILI has to offer ADSC therapy in regenerative medicine.


Subject(s)
Adipocytes/radiation effects , Cell Differentiation/radiation effects , Low-Level Light Therapy/methods , Mesenchymal Stem Cells/radiation effects , Myocytes, Smooth Muscle/radiation effects , Adipocytes/physiology , Adipose Tissue/cytology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Coculture Techniques/methods , Humans , Mesenchymal Stem Cells/physiology , Myocytes, Smooth Muscle/physiology , Role , Sensitivity and Specificity
13.
J Mol Cell Cardiol ; 81: 62-70, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640160

ABSTRACT

Age-associated cardiovascular diseases are at least partially ascribable to vascular cell senescence. Replicative senescence (RS) and stress-induced premature senescence (SIPS) are provoked respectively by endogenous (telomere erosion) and exogenous (H2O2, UV) stimuli resulting in cell cycle arrest in G1 and G2 phases. In both scenarios, mitochondria-derived ROS are important players in senescence initiation. We aimed to define whether a mtDNA-transcribed long-non-coding-RNA (lncRNA), ASncmtRNA-2, has a role in vascular aging and senescence. Aortas of old mice, characterized by increased senescence, showed an increment in ASncmtRNA-2 expression. In vitro analysis of Endothelial Cells (EC) and Vascular Smooth Muscle Cells (VSMC) established that ASncmtRNA-2 is induced in EC, but not in VSMC, during RS. Surprisingly, ASncmtRNA-2 is not upregulated in two different EC SIPS scenarios, treated with H2O2 and UV. The p16 gene displayed similar ASncmtRNA-2 expression patterns, suggesting a possible co-regulation of the two genes. Interestingly, the expression of two miRNAs, hsa-miR-4485 and hsa-miR-1973, with perfect homology to the double strand region of ASncmtRNA-2 and originating at least in part from a mitochondrial transcript, was induced in RS, opening to the possibility that this lncRNA functions as a non-canonical precursor of these miRNAs. Cell cycle analysis of EC transiently over-expressing ASncmtRNA-2 revealed an accumulation of EC in the G2/M phase, but not in the G1 phase. We propose that ASncmtRNA-2 in EC might be involved in the RS establishment by participating in the cell cycle arrest in G2/M phase, possibly through the production of hsa-miR-4485 and hsa-miR-1973. This article is part of a Special Issue entitled: Mitochondria.


Subject(s)
Aging/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , RNA, Long Noncoding/genetics , RNA/genetics , Aging/genetics , Animals , Aorta/cytology , Aorta/metabolism , Base Sequence , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Molecular Sequence Data , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/radiation effects , RNA/metabolism , RNA, Long Noncoding/metabolism , RNA, Mitochondrial , Signal Transduction , Ultraviolet Rays
14.
Toxicol Sci ; 144(1): 138-50, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25516496

ABSTRACT

Because of high exposure to systemic noxae, vascular endothelial cells (EC) have to ensure distinct damage defense and regenerative mechanisms to guarantee vascular health. For meaningful toxicological drug assessments employing embryonic stem cell (ESC)-based in vitro models, functional competence of differentiated progeny and detailed knowledge regarding damage defense mechanisms are essential. Here, mouse ESCs (mESC) were differentiated into functionally competent vascular cells (EC and smooth muscle cells [SMC]). mESC, EC, and SMC were comparatively analyzed regarding DNA repair and DNA damage response (DDR). Differentiation was accompanied by both congruent and unique alterations in repair and DDR characteristics. EC and SMC shared the downregulation of genes involved cell cycle regulation and repair of DNA double-strand breaks (DSBs) and mismatches, whereas genes associated with nucleotide excision repair (NER), apoptosis, and autophagy were upregulated when compared with mESC. Expression of genes involved in base excision repair (BER) was particularly low in SMC. IR-induced formation of DSBs, as detected by nuclear γH2AX foci formation, was most efficient in SMC, the repair of DSBs was fastest in EC. Together with substantial differences in IR-induced phosphorylation of p53, Chk1, and Kap1, the data demonstrate complex alterations in DDR capacity going along with the loss of pluripotency and gain of EC- and SMC-specific functions. Notably, IR exposure of early vascular progenitors did not impair differentiation into functionally competent EC and SMC. Summarizing, mESC-based vascular differentiation models are informative to study the impact of environmental stressors on differentiation and function of vascular cells.


Subject(s)
Cell Differentiation/radiation effects , Embryonic Stem Cells/radiation effects , Endothelial Progenitor Cells/radiation effects , Muscle, Smooth, Vascular/radiation effects , Myocytes, Smooth Muscle/radiation effects , Pluripotent Stem Cells/radiation effects , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Autophagy/radiation effects , Biomarkers/metabolism , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA Breaks, Double-Stranded , DNA Repair , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Gene Expression Regulation , Histones/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/radiation effects , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , RNA, Messenger/metabolism , Time Factors
15.
Genet Mol Res ; 13(2): 3143-53, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24782171

ABSTRACT

Ultrasound, a non-invasive therapy method, is a potential tool for medical applications, but its biological effects on vascular smooth muscle cells (VSMCs) have not been characterized. The aim of this study was to explore the effect and possible apoptotic mechanism of VSMCs that were induced by low-frequency ultrasound (LFU). Cell viability and apoptosis of A7r5 cells were evaluated after treating A7r5 cells with a continuous 45-kHz 1.0-W/cm(2) ultrasound (exposure time of 0, 10, 20, 30, and 35 s) by MTT assay and flow cytometry. At the optimum ultrasound exposure condition (30 s), gene chip analysis was performed, and the apoptotic signaling pathway was confirmed by reverse transcription-polymerase chain reaction and Western blot. As measured by flow cytometry, LFU significantly induced A7r5 cell apoptosis. Comparing the ultrasound group with the control group, the protein expression of caspase-9 and caspase-3 was increased by 50 and 57%, respectively; the caspase-3 mRNA level was increased by 37.5%. These findings indicate that an intrinsic pathway plays a major role in apoptosis that is induced by LFU and that LFU can induce A7r5 cell apoptosis via caspase-9- and caspase-3-dependent pathways.


Subject(s)
Aorta/metabolism , Apoptosis/radiation effects , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/cytology , Aorta/radiation effects , Caspase 3/biosynthesis , Caspase 9/biosynthesis , Cell Line , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Gene Expression Regulation, Enzymologic/radiation effects , Myocytes, Smooth Muscle/radiation effects , RNA, Messenger/biosynthesis , Rats , Sound
16.
Int J Radiat Biol ; 90(10): 914-27, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24713030

ABSTRACT

PURPOSE: γ-rays (IR) cause an increase in intracellular calcium [Ca(2+)], alters contractility and triggers apoptosis via the activation of protein kinase C in intestinal guinea pig smooth muscle cells. The present study investigated the role of the mitochondria in these processes and characterized proteins involved in IR-induced apoptosis. MATERIALS AND METHODS: Intestinal smooth muscle cells were exposed to 10-50 Gy from a (60)Co γ-source. Reactive oxygen species (ROS) levels were measured by colourimetry with a fluorescente probe. Protein expression was analyzed by immunoblotting and immunofluorescence. RESULTS: Apoptosis was inhibited by glutathione, possible by inhibiting the generation or scavenging ROS. Apoptosis was mediated by the mitochondria releasing cytochrome c leading to caspase 3 activation. IR increased the expression of the cyclins A, B2 and E and led to unbalanced cellular growth in an absorption dose-dependent manner. However, radiation did not induce alterations in the mitochondrial ultrastructure or in transmembrane electric potential. In contrast, IR increased the nuclear expression of cytoplasmic proteins and cyclins A and E. CONCLUSION: Smooth muscle cells subjected to IR undergo mitochondrial-mediated apoptosis that involves oncoproteins activation and preserves mitochondrial structure. IR also cause alterations in the expression and localization of both pro- and anti-apoptotic proteins.


Subject(s)
Apoptosis/physiology , Calcium Signaling/physiology , Mitochondria, Muscle/physiology , Muscle Contraction/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Calcium/metabolism , Calcium Signaling/radiation effects , Cell Cycle/physiology , Cell Cycle/radiation effects , Cell Cycle Proteins/metabolism , Cells, Cultured , Dose-Response Relationship, Radiation , Gamma Rays , Guinea Pigs , Mitochondria, Muscle/radiation effects , Muscle Contraction/radiation effects , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/radiation effects , Radiation Dosage
17.
Ultrasound Med Biol ; 39(7): 1267-76, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23643062

ABSTRACT

Focal drug delivery to a vessel wall facilitated by intravascular ultrasound and microbubbles holds promise as a potential therapy for atherosclerosis. Conventional methods of microbubble administration result in rapid clearance from the bloodstream and significant drug loss. To address these limitations, we evaluated whether drug delivery could be achieved with transiently stable microbubbles produced in real time and in close proximity to the therapeutic site. Rat aortic smooth muscle cells were placed in a flow chamber designed to simulate physiological flow conditions. A flow-focusing microfluidic device produced 8 µm diameter monodisperse microbubbles within the flow chamber, and ultrasound was applied to enhance uptake of a surrogate drug (calcein). Acoustic pressures up to 300 kPa and flow rates up to 18 mL/s were investigated. Microbubbles generated by the flow-focusing microfluidic device were stabilized with a polyethylene glycol-40 stearate shell and had either a perfluorobutane (PFB) or nitrogen gas core. The gas core composition affected stability, with PFB and nitrogen microbubbles exhibiting half-lives of 40.7 and 18.2 s, respectively. Calcein uptake was observed at lower acoustic pressures with nitrogen microbubbles (100 kPa) than with PFB microbubbles (200 kPa) (p < 0.05, n > 3). In addition, delivery was observed at all flow rates, with maximal delivery (>70% of cells) occurring at a flow rate of 9 mL/s. These results demonstrate the potential of transiently stable microbubbles produced in real time and in close proximity to the intended therapeutic site for enhancing localized drug delivery.


Subject(s)
Drug Delivery Systems/instrumentation , Fluoresceins/pharmacokinetics , Microbubbles/therapeutic use , Microfluidic Analytical Techniques/instrumentation , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/radiation effects , Sonication/instrumentation , Animals , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Rats
18.
Opt Lett ; 37(24): 5030-2, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23257994

ABSTRACT

Methods of controllable, noncontact rotation of optically trapped microscopic objects have garnered significant attention for tomographic imaging and microfluidic actuation. Here, we report development of a fiber-optic spanner and demonstrate controlled rotation of smooth muscle cells. The rotation is realized by introducing a transverse offset between two counterpropagating beams emanating from single-mode optical fibers. The rotation speed and surrounding microfluidic flow could be controlled by varying balanced laser beam powers. Further, we demonstrate simultaneous translation and rotation of the fiber-optically trapped cell by varying the laser power of one fiber-optic arm.


Subject(s)
Cell Separation/instrumentation , Cell Tracking/instrumentation , Fiber Optic Technology/instrumentation , Microfluidic Analytical Techniques/instrumentation , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Optical Tweezers , Cell Movement/physiology , Cell Polarity , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Humans , Light , Myocytes, Smooth Muscle/radiation effects , Rotation
19.
Semin Dial ; 25(4): 464-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22276964

ABSTRACT

Hemodialysis vascular access dysfunction is a major cause of morbidity and hospitalization in the hemodialysis population at a cost of well over $1 billion per annum. Venous stenosis (due to venous neointimal hyperplasia [VNH]) is the most common cause of polytetrafluroethylene PTFE) dialysis access graft and arteriovenous fistula (AVF) failure. Despite the magnitude of the clinical problem, however, there are currently no effective therapies for this condition. We and others have previously demonstrated that VNH in PTFE dialysis grafts and AVF is composed of smooth muscle cells/myofibroblasts, endothelial cells within neointimal microvessels, and peri-graft macrophages. Radiation therapy blocks the proliferation and activation of all these cell types. The current review will dissect out the available in vitro, experimental, and clinical data on the use of radiation therapy for vascular stenosis in general, and for dialysis access dysfunction in particular. It is important to try and identify whether there is still a role for radiation therapy in this specific clinical setting. We believe that this is a critically important question to answer in view of the huge unmet clinical need that is currently associated with hemodialysis vascular access dysfunction.


Subject(s)
Arteriovenous Shunt, Surgical , Graft Occlusion, Vascular/radiotherapy , Renal Dialysis , Animals , Beta Particles , Blood Vessel Prosthesis , Brachytherapy , Cell Proliferation , Drug-Eluting Stents , Endothelial Cells/radiation effects , Gamma Rays , Graft Occlusion, Vascular/etiology , Humans , Hyperplasia/etiology , Hyperplasia/radiotherapy , Kidney Failure, Chronic/therapy , Macrophages/radiation effects , Myocytes, Smooth Muscle/radiation effects , Myofibroblasts/radiation effects , Polytetrafluoroethylene , Radiotherapy Dosage , Tunica Intima/pathology , Veins/pathology
20.
J Surg Res ; 176(2): 376-85, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22172131

ABSTRACT

BACKGROUND: The extracellular matrix plays an important role in heart valve function. To improve the processing of porcine pulmonary valves for clinical use, we have studied the influence of cryopreservation, decellularization, and irradiation on extracellular matrix components. METHODS: Decellularization was carried out followed by DNAseI/RNAseA digestion and isotonic washout. Valves were cryopreserved in 10% DMSO/10% fetal bovine serum, and then subjected to 25-40 kGy γ-radiation. Extracellular matrix constituents were evaluated by histologic staining, immunohistochemistry, transmission electron microscopy, and liquid chromatography/mass spectrometry. RESULTS: Histologic, immunohistochemical, ultrastructural, and biochemical analyses demonstrated a marked reduction in the expression of extracellular matrix components particularly in the valves that had been γ-irradiated following decellularization and cryopreservation. In this group, histology and immunohistochemistry showed an obvious reduction in staining for chondroitin sulphates, versican, hyaluronan, and collagens. Transmission electron microscopy revealed the smallest fibril diameter of collagen, shortest D-period, and loss of compactness of collagen fiber packaging and fragmentation of elastic fibers. Biochemical analysis showed loss of collagen and elastin crosslinks. Decellularization followed by cryopreservation showed some reduction in staining for collagens and versican, smaller diameter, shorter D-period in collagen fibers, and ridges in elastic fibers. Cryopreservation alone showed minimal changes in ECM staining intensity, collagen, and elastin ultrastructure and biochemistry. CONCLUSION: γ-Irradiated valves that have been decellularized and cryopreserved produces significant changes in the expression of ECM components, thus providing useful information for improving valve preparation for clinical use and also some indication as to why irradiated human heart valves were not clinically successful.


Subject(s)
Cryopreservation/methods , Extracellular Matrix/radiation effects , Gamma Rays/adverse effects , Pulmonary Valve/radiation effects , Pulmonary Valve/transplantation , Animals , Collagen/metabolism , Coronary Sinus/radiation effects , Coronary Sinus/ultrastructure , Cross-Linking Reagents/metabolism , Elastin/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Glycosaminoglycans/metabolism , Humans , Mass Spectrometry , Microscopy, Electron, Transmission , Myocytes, Smooth Muscle/radiation effects , Myocytes, Smooth Muscle/ultrastructure , Pulmonary Valve/ultrastructure , Swine , Transplantation, Heterologous , Versicans/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...