Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-34829990

RESUMO

Low-intensity pulsed ultrasound (LIPUS) has been proved to promote the proliferation of myoblast C2C12. However, whether LIPUS can effectively prevent muscle atrophy has not been clarified, and if so, what is the possible mechanism. The aim of this study is to evaluate the effects of LIPUS on muscle atrophy in hindlimb unloading rats, and explore the mechanisms. The rats were randomly divided into four groups: normal control group (NC), hindlimb unloading group (UL), hindlimb unloading plus 30 mW/cm2 LIPUS irradiation group (UL + 30 mW/cm2), hindlimb unloading plus 80 mW/cm2 LIPUS irradiation group (UL + 80 mW/cm2). The tails of rats in hindlimb unloading group were suspended for 28 days. The rats in the LIPUS treated group were simultaneously irradiated with LIPUS on gastrocnemius muscle in both lower legs at the sound intensity of 30 mW/cm2 or 80 mW/cm2 for 20 min/d for 28 days. C2C12 cells were exposed to LIPUS at 30 or 80 mW/cm2 for 5 days. The results showed that LIPUS significantly promoted the proliferation and differentiation of myoblast C2C12, and prevented the decrease of cross-sectional area of muscle fiber and gastrocnemius mass in hindlimb unloading rats. LIPUS also significantly down regulated the expression of MSTN and its receptors ActRIIB, and up-regulated the expression of Akt and mTOR in gastrocnemius muscle of hindlimb unloading rats. In addition, three metabolic pathways (phenylalanine, tyrosine and tryptophan biosynthesis; alanine, aspartate and glutamate metabolism; glycine, serine and threonine metabolism) were selected as important metabolic pathways for hindlimb unloading effect. However, LIPUS promoted the stability of alanine, aspartate and glutamate metabolism pathway. These results suggest that the key mechanism of LIPUS in preventing muscle atrophy induced by hindlimb unloading may be related to promoting protein synthesis through MSTN/Akt/mTOR signaling pathway and stabilizing alanine, aspartate and glutamate metabolism.


Assuntos
Diferenciação Celular/efeitos da radiação , Atrofia Muscular/terapia , Ondas Ultrassônicas , Receptores de Activinas Tipo II/genética , Animais , Proliferação de Células/genética , Proliferação de Células/efeitos da radiação , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos da radiação , Membro Posterior/patologia , Membro Posterior/efeitos da radiação , Elevação dos Membros Posteriores/métodos , Humanos , Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/efeitos da radiação , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Músculo Esquelético/efeitos da radiação , Atrofia Muscular/genética , Atrofia Muscular/patologia , Mioblastos/efeitos da radiação , Miostatina/genética , Ratos , Terapia por Ultrassom/métodos
2.
Clin Orthop Relat Res ; 479(3): 468-474, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33252888

RESUMO

BACKGROUND: Radiation-induced fibrosis is a long-term adverse effect of external beam radiation therapy for cancer treatment that can cause pain, loss of function, and decreased quality of life. Transforming growth factor beta (TGF-ß) is believed to be critical to the development of radiation-induced fibrosis, and TGF-ß inhibition decreases the development of fibrosis. However, no treatment exists to prevent radiation-induced fibrosis. Therefore, we aimed to mitigate the development of radiation-induced fibrosis in a mouse model by inhibiting TGF-ß. QUESTION/PURPOSES: Does TGF-ß inhibition decrease the development of muscle fibrosis induced by external beam radiation in a mouse model? METHODS: Twenty-eight 12-week-old male C57BL/6 mice were assigned randomly to three groups: irradiated mice treated with TGF-ßi, irradiated mice treated with placebo, and control mice that received neither irradiation nor treatment. The irradiated mice received one 50-Gy fraction of radiation to the right hindlimb before treatment initiation. Mice treated with TGF-c (n = 10) received daily intraperitoneal injections of a small-molecule inhibitor of TGF-ß (1 mg/kg) in a dimethyl sulfoxide vehicle for 8 weeks (seven survived to histologic analysis). Mice treated with placebo (n = 10) received daily intraperitoneal injections of only a dimethyl sulfoxide vehicle for 8 weeks (10 survived to histologic analysis). Control mice (n = 8) received neither radiation nor TGF-ß treatment. Control mice were euthanized at 3 months because they were not expected to exhibit any changes related to treatment. Mice in the two treatment groups were euthanized 9 months after radiation, and the quadriceps of each thigh was sampled. Masson's trichome stain was used to assess muscle fibrosis. Slides were viewed at 10 × magnification using bright-field microscopy, and in a blinded fashion, five representative images per mouse were used to quantify fibrosis. The mean ± SD fibrosis pixel densities in the TGF-ßi and radiation-only groups were compared using Mann-Whitney U tests. The ratio of fibrosis to muscle was calculated using the mean fibrosis per slide in the TGF-ßi group to standardize measurements. Alpha was set at 0.05. RESULTS: The mean (± SD) percentage of fibrosis per slide was greater in the radiation-only group (1.2% ± 0.42%) than in the TGF-ßi group (0.14% ± 0.09%) (odds ratio 0.12 [95% CI 0.07 to 0.20]; p < 0.001). Among control mice, mean fibrosis was 0.05% ± 0.02% per slide. Mice in the radiation-only group had 9.1 times the density of fibrosis as did mice in the TGF-ßi group. CONCLUSION: Our study provides preliminary evidence that the fibrosis associated with radiation therapy to a quadriceps muscle can be reduced by treatment with a TGF-ß inhibitor in a mouse model. CLINICAL RELEVANCE: If these observations are substantiated by further investigation into the role of TGF-ß inhibition on the development of radiation-induced fibrosis in larger animal models and humans, our results may aid in the development of novel therapies to mitigate this complication of radiation treatment.


Assuntos
Membro Posterior/patologia , Músculo Quadríceps/patologia , Lesões por Radiação/prevenção & controle , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Fibrose , Membro Posterior/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Quadríceps/efeitos da radiação , Lesões por Radiação/patologia
3.
Sci Rep ; 10(1): 19501, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33177579

RESUMO

Pediatric cancer treatment often involves chemotherapy and radiation, where off-target effects can include skeletal muscle decline. The effect of such treatments on juvenile skeletal muscle growth has yet to be investigated. We employed a small animal irradiator to administer fractionated hindlimb irradiation to juvenile mice bearing implanted rhabdomyosarcoma (RMS) tumors. Hindlimb-targeted irradiation (3 × 8.2 Gy) of 4-week-old mice successfully eliminated RMS tumors implanted one week prior. After establishment of this preclinical model, a cohort of tumor-bearing mice were injected with the chemotherapeutic drug, vincristine, alone or in combination with fractionated irradiation (5 × 4.8 Gy). Single myofiber analysis of fast-contracting extensor digitorum longus (EDL) and slow-contracting soleus (SOL) muscles was conducted 3 weeks post-treatment. Although a reduction in myofiber size was apparent, EDL and SOL myonuclear number were differentially affected by juvenile irradiation and/or vincristine treatment. In contrast, a decrease in myonuclear domain (myofiber volume/myonucleus) was observed regardless of muscle or treatment. Thus, inhibition of myofiber hypertrophic growth is a consistent feature of pediatric cancer treatment.


Assuntos
Quimiorradioterapia/efeitos adversos , Fibras Musculares Esqueléticas/patologia , Rabdomiossarcoma/terapia , Envelhecimento , Animais , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fracionamento da Dose de Radiação , Membro Posterior/efeitos dos fármacos , Membro Posterior/patologia , Membro Posterior/efeitos da radiação , Hipertrofia , Masculino , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos da radiação , Teste de Desempenho do Rota-Rod , Transplante Isogênico , Vincristina/farmacologia
4.
Ultrasound Med Biol ; 46(9): 2361-2369, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32522456

RESUMO

Intra-vascular ultrasound catheters are used clinically to facilitate clot lysis. We hypothesized that these devices could also directly lower microvascular resistance and increase tissue perfusion through established shear-dependent pathways. In mice, either the proximal hind-limb muscles or the upstream femoral artery alone was exposed to an endovascular ultrasound catheter (2.3 MHz, 0.5-1.1 MPa) for 10 min. Quantitative microvascular perfusion imaging in the hind limbs exposed to the endovascular ultrasound system exhibited a more-than-twofold increase in flow (p < 0.01) compared with the contralateral control limb after exposure of either the muscle or the femoral artery alone. Using an in vivo optical imaging reporting system, an eight- to ninefold increase in tissue adenosine triphosphate (ATP) was detected in the region of insonification (p = 0.006). Ultrasound was found to produce an immediate release of ATP from ex vivo erythrocytes (p = 0.03). In situ electrochemical sensing revealed an immediate increase in nitric oxide with initiation of ultrasound which returned to baseline within 5 min of termination, as well as ultrasound-triggered nitric oxide (NO) release from erythrocytes. These data indicate that non-cavitating ultrasound produced by endovascular catheters can reduce vascular resistance and increase flow through recognized shear-dependent vasodilator pathways involving purinergic signaling and NO.


Assuntos
Catéteres , Endossonografia/instrumentação , Artéria Femoral/fisiologia , Artéria Femoral/efeitos da radiação , Membro Posterior/irrigação sanguínea , Membro Posterior/efeitos da radiação , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos da radiação , Fluxo Sanguíneo Regional , Ultrassonografia de Intervenção/instrumentação , Animais , Camundongos , Camundongos Endogâmicos C57BL , Resistência Vascular/efeitos da radiação
5.
J Biomed Opt ; 23(10): 1-9, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30315644

RESUMO

Radiation therapy (RT) is widely and effectively used for cancer treatment but can also cause deleterious side effects, such as a late-toxicity complication called radiation-induced fibrosis (RIF). Accurate diagnosis of RIF requires analysis of histological sections to assess extracellular matrix infiltration. This is invasive, prone to sampling limitations, and thus rarely used; instead, current practice relies on subjective clinical surrogates, including visual observation, palpation, and patient symptomatology questionnaires. This preclinical study demonstrates that functional optical coherence tomography (OCT) is a useful tool for objective noninvasive in-vivo assessment and quantification of fibrosis-associated microvascular changes in tissue. Data were collected from murine hind limbs 6 months after 40-Gy single-dose irradiation and compared with nonirradiated contralateral tissues of the same animals. OCT-derived vascular density and average vessel diameter metrics were compared to quantitative vascular analysis of stained histological slides. Results indicate that RIF manifests significant microvascular changes at this time point posttreatment. Abnormal microvascular changes visualized by OCT in this preclinical setting suggest the potential of this label-free high-resolution noninvasive functional imaging methodology for RIF diagnosis and assessment in the context of clinical RT.


Assuntos
Fibrose/diagnóstico por imagem , Microcirculação/efeitos da radiação , Radioterapia/efeitos adversos , Pele , Tomografia de Coerência Óptica/métodos , Animais , Feminino , Membro Posterior/irrigação sanguínea , Membro Posterior/diagnóstico por imagem , Membro Posterior/efeitos da radiação , Camundongos , Camundongos Endogâmicos C3H , Lesões Experimentais por Radiação/diagnóstico por imagem , Pele/irrigação sanguínea , Pele/diagnóstico por imagem , Pele/efeitos da radiação
6.
Bioelectromagnetics ; 39(8): 569-584, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30350869

RESUMO

Microgravity is one of the main threats to the health of astronauts. Pulsed electromagnetic fields (PEMFs) have been considered as one of the potential countermeasures for bone loss induced by space flight. However, the optimal therapeutic parameters of PEMFs have not been obtained and the action mechanism is still largely unknown. In this study, a set of optimal therapeutic parameters for PEMFs (50 Hz, 0.6 mT 50% duty cycle and 90 min/day) selected based on high-throughput screening with cultured osteoblasts was used to prevent bone loss in rats induced by hindlimb suspension, a commonly accepted animal model to simulate the space environment. It was found that hindlimb suspension for 4 weeks led to significant decreases in femoral and vertebral bone mineral density (BMD) and their maximal loads, severe deterioration in bone micro-structure, and decreases in levels of bone formation markers and increases in bone resorption markers. PEMF treatment prevented about 50% of the decreased BMD and maximal loads, preserved the microstructure of cancellous bone and thickness of cortical bone, and inhibited decreases in bone formation markers. Histological analyses revealed that PEMFs significantly alleviated the reduction in osteoblast number and inhibited the increase in adipocyte number in the bone marrow. PEMFs also blocked decreases in serum levels of parathyroid hormone and its downstream signal molecule cAMP, and maintained the phosphorylation levels of protein kinase A (PKA) and cAMP response element-binding protein (CREB). The expression level of soluble adenylyl cyclases (sAC) was also maintained. It therefore can be concluded that PEMFs partially prevented the bone loss induced by weightless environment by maintaining bone formation through signaling of the sAC/cAMP/PKA/CREB pathway. Bioelectromagnetics. 39:569-584, 2018. © 2018 Wiley Periodicals, Inc.


Assuntos
Adenilil Ciclases/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Campos Eletromagnéticos , Membro Posterior/fisiologia , Osteogênese/efeitos da radiação , Adipócitos/citologia , Adipócitos/efeitos da radiação , Animais , Fenômenos Biomecânicos/efeitos da radiação , Peso Corporal/efeitos da radiação , Densidade Óssea/efeitos da radiação , Reabsorção Óssea/metabolismo , Reabsorção Óssea/prevenção & controle , Feminino , Fêmur/citologia , Fêmur/diagnóstico por imagem , Fêmur/fisiologia , Fêmur/efeitos da radiação , Membro Posterior/efeitos da radiação , Osteoblastos/citologia , Osteoblastos/efeitos da radiação , Ratos , Ratos Wistar , Transdução de Sinais/efeitos da radiação , Coluna Vertebral/citologia , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/fisiologia , Coluna Vertebral/efeitos da radiação , Suspensões , Microtomografia por Raio-X
7.
J Bone Miner Res ; 33(1): 99-112, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28902435

RESUMO

Radiation therapy (RTx) is associated with increased risk for late-onset fragility fractures in bone tissue underlying the radiation field. Bone tissue outside the RTx field is often selected as a "normal" comparator tissue in clinical assessment of fragility fracture risk, but the robustness of this comparison is limited by an incomplete understanding of the systemic effects of local radiotherapy. In this study, a mouse model of limited field irradiation was used to quantify longitudinal changes in local (irradiated) and systemic (non-irradiated) femurs with respect to bone density, morphology, and strength. BALB/cJ mice aged 12 weeks underwent unilateral hindlimb irradiation (4 × 5 Gy) or a sham procedure. Femurs were collected at endpoints of 4 days before treatment and at 0, 1, 2, 4, 8, 12, and 26 weeks post-treatment. Irradiated (RTx), Contralateral (non-RTx), and Sham (non-RTx) femurs were imaged by micro-computed tomography and mechanically tested in three-point bending. In both the RTx and Contralateral non-RTx groups, the longer-term (12- to 26-week) outcomes included trabecular resorption, loss of diaphyseal cortical bone, and decreased bending strength. Contralateral femurs generally followed an intermediate response compared with RTx femurs. Change also varied by anatomic compartment; post-RTx loss of trabecular bone was more profound in the metaphyseal than the epiphyseal compartment, and cortical bone thickness decreased at the mid-diaphysis but increased at the metaphysis. These data demonstrate that changes in bone quantity, density, and architecture occur both locally and systemically after limited field irradiation and vary by anatomic compartment. Furthermore, the severity and persistence of systemic bone damage after limited field irradiation suggest selection of control tissues for assessment of fracture risk or changes in bone density after radiotherapy may be challenging. © 2017 American Society for Bone and Mineral Research.


Assuntos
Fêmur/anatomia & histologia , Fêmur/fisiologia , Membro Posterior/efeitos da radiação , Animais , Fenômenos Biomecânicos , Densidade Óssea/efeitos da radiação , Osso Esponjoso/anatomia & histologia , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/fisiologia , Osso Esponjoso/efeitos da radiação , Osso Cortical/anatomia & histologia , Osso Cortical/diagnóstico por imagem , Osso Cortical/fisiologia , Osso Cortical/efeitos da radiação , Diáfises/anatomia & histologia , Diáfises/diagnóstico por imagem , Diáfises/fisiologia , Diáfises/efeitos da radiação , Feminino , Fêmur/diagnóstico por imagem , Fêmur/efeitos da radiação , Estudos Longitudinais , Camundongos Endogâmicos BALB C , Microtomografia por Raio-X
8.
Bioelectromagnetics ; 39(2): 89-97, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29251361

RESUMO

Pulsed electromagnetic field (PEMF) is often used for management of osteoarthritis (OA). The aim of the study was to determine whether PEMF can successfully improve subchondral bone microstructure through a Wnt/ß-catenin signaling-associated pathway in rats with knee OA induced by low-dose monosodium iodoacetate (MIA). Seventy-two 12-week-old male Sprague-Dawley rats were randomly assigned to three groups: OA (n = 24), PEMF (n = 24), and Control (n = 24). OA was induced (OA and PEMF groups) by injecting 0.2 mg MIA in rats' right knee joint. The control rats received a single sterile saline injection in the right knee. Rats in the PEMF group were exposed to daily 2 h PEMF exposure with 75 Hz, 1.6 mT for 4 weeks. After 4 weeks, micro-computed tomography (micro-CT), real-time PCR, and immunohistochemistry staining were performed. The PEMF group increased bone volume fraction (BV/TV), trabecular thickness (Tb.Th), trabecular number (Tb.N), and suppressed bone surface/bone volume (BS/BV) and trabecular separation (Tb.Sp) levels in micro-CT analysis. Real-time PCR analysis showed that PEMF promoted tibial subchondral bone's gene expressions of Wnt3a, ß-catenin, and OPG, but did not alter LRP5 and RANKL mRNA levels. Similar results involved tibial subchondral bone's protein expressions that were observed in immunohistochemistry staining. These results suggest that PEMF preserved the structural integrity of subchondral bone in knee OA rats by promoting the activation of Wnt/ß-catenin signaling and OPG/RANKL/RANK signaling. Bioelectromagnetics. 39:89-97, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Membro Posterior/patologia , Membro Posterior/efeitos da radiação , Magnetoterapia , Osteoartrite do Joelho/patologia , Via de Sinalização Wnt/efeitos da radiação , Animais , Membro Posterior/diagnóstico por imagem , Membro Posterior/metabolismo , Masculino , Osteoartrite do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/metabolismo , Ratos , Ratos Sprague-Dawley , Microtomografia por Raio-X
9.
Plast Reconstr Surg ; 137(5): 1486-1497, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27119923

RESUMO

BACKGROUND: Treatment of critical limb ischemia is sometimes difficult because of the patient's condition, and some novel approaches are needed. METHODS: The hindlimbs of Sprague-Dawley rats, after 20-Gy x-ray irradiation and surgical occlusion, were divided into four groups: with a superficial fascial flap, 5.0 × 10 adipose-derived stromal/stem cells, and both combined. The rats were tested for laser tissue blood flow, immunohistologic blood vessel density, and foot paw punch hole wound healing. Green fluorescent protein-tagged Sprague-Dawley rats were used for further investigation by cell tracking for 2 weeks. RESULTS: Laser tissue blood flow demonstrated a significant increase in the combined treatment of flap and adipose-derived stem cells at both 1 and 2 weeks. There were no significant differences between the treatment groups treated with flaps alone and those treated with adipose-derived stem cells alone. Wound healing was significantly increased following combined treatment at 1 week, and there was no wound by 2 weeks except for the no-flap and no-adipose-derived stem cell group. The number of vessels depicted by von Willebrand factor showed a significant increase in the combined treatment group, at both 1 week and 2 weeks. In the cell tracking group, at 2 weeks, the green fluorescent protein-tagged adipose-derived stem cells were significantly more positive in the no-flap group than in the flap group. CONCLUSIONS: Adipose-derived stem cells may be a potent cell source in irradiated and occluded limbs by enhancing tissue blood flow and blood vessel density. Adipose-derived stem cells may play an important role in some difficult ischemic conditions in terms of wound healing.


Assuntos
Tecido Adiposo/citologia , Isquemia/cirurgia , Neovascularização Fisiológica , Transplante de Células-Tronco , Retalhos Cirúrgicos , Cicatrização , Animais , Proteínas de Fluorescência Verde/análise , Membro Posterior/irrigação sanguínea , Membro Posterior/efeitos da radiação , Isquemia/terapia , Masculino , Lesões Experimentais por Radiação/cirurgia , Lesões Experimentais por Radiação/terapia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Retalhos Cirúrgicos/irrigação sanguínea , Fator de von Willebrand/análise
10.
Radiat Res ; 185(3): 257-66, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26930379

RESUMO

Weightlessness during spaceflight leads to functional changes in resistance arteries and loss of cancellous bone, which may be potentiated by radiation exposure. The purpose of this study was to assess the effects of hindlimb unloading (HU) and total-body irradiation (TBI) on the vasomotor responses of skeletal muscle arteries. Male C57BL/6 mice were assigned to control, HU (13-16 days), TBI (1 Gy (56)Fe, 600 MeV, 10 cGy/min) and HU-TBI groups. Gastrocnemius muscle feed arteries were isolated for in vitro study. Endothelium-dependent (acetylcholine) and -independent (Dea-NONOate) vasodilator and vasoconstrictor (KCl, phenylephrine and myogenic) responses were evaluated. Arterial endothelial nitric oxide synthase (eNOS), superoxide dismutase-1 (SOD-1) and xanthine oxidase (XO) protein content and tibial cancellous bone microarchitecture were quantified. Endothelium-dependent and -independent vasodilator responses were impaired in all groups relative to control, and acetylcholine-induced vasodilation was lower in the HU-TBI group relative to that in the HU and TBI groups. Reductions in endothelium-dependent vasodilation correlated with a lower cancellous bone volume fraction. Nitric oxide synthase inhibition abolished all group differences in endothelium-dependent vasodilation. HU and HU-TBI resulted in decreases in eNOS protein levels, while TBI and HU-TBI produced lower SOD-1 and higher XO protein content. Vasoconstrictor responses were not altered. Reductions in NO bioavailability (eNOS), lower anti-oxidant capacity (SOD-1) and higher pro-oxidant capacity (XO) may contribute to the deficits in NOS signaling in skeletal muscle resistance arteries. These findings suggest that the combination of insults experienced in spaceflight leads to impairment of vasodilator function in resistance arteries that is mediated through deficits in NOS signaling.


Assuntos
Músculo Esquelético/efeitos da radiação , Exposição à Radiação , Vasodilatação/efeitos da radiação , Sistema Vasomotor/efeitos da radiação , Animais , Artérias/metabolismo , Artérias/efeitos da radiação , Membro Posterior/metabolismo , Membro Posterior/efeitos da radiação , Humanos , Masculino , Camundongos , Músculo Esquelético/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Voo Espacial , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Vasodilatadores/administração & dosagem , Sistema Vasomotor/metabolismo , Irradiação Corporal Total , Xantina Oxidase/metabolismo
11.
Proc Natl Acad Sci U S A ; 112(32): 9920-5, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26216955

RESUMO

The postnatal skeleton undergoes growth, remodeling, and repair. We hypothesized that skeletal progenitor cells active during these disparate phases are genetically and phenotypically distinct. We identified a highly potent regenerative cell type that we term the fracture-induced bone, cartilage, stromal progenitor (f-BCSP) in the fracture callus of adult mice. The f-BCSP possesses significantly enhanced skeletogenic potential compared with BCSPs harvested from uninjured bone. It also recapitulates many gene expression patterns involved in perinatal skeletogenesis. Our results indicate that the skeletal progenitor population is functionally stratified, containing distinct subsets responsible for growth, regeneration, and repair. Furthermore, our findings suggest that injury-induced changes to the skeletal stem and progenitor microenvironments could activate these cells and enhance their regenerative potential.


Assuntos
Osso e Ossos/patologia , Fraturas Ósseas/patologia , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Desenvolvimento Ósseo , Calo Ósseo/citologia , Cartilagem/patologia , Proliferação de Células , Separação Celular , Fêmur/patologia , Perfilação da Expressão Gênica , Membro Posterior/efeitos da radiação , Integrina alfa6/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Osteogênese , Fenótipo , Células Estromais/citologia
12.
J Bone Miner Res ; 30(7): 1268-79, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25588731

RESUMO

Increased fracture risk is commonly reported in cancer patients receiving radiotherapy, particularly at sites within the field of treatment. The direct and systemic effects of ionizing radiation on bone at a therapeutic dose are not well-characterized in clinically relevant animal models. Using 20-week-old male C57Bl/6 mice, effects of irradiation (right hindlimb; 2 Gy) on bone volume and microarchitecture were evaluated prospectively by microcomputed tomography and histomorphometry and compared to contralateral-shielded bone (left hindlimb) and non-irradiated control bone. One week postirradiation, trabecular bone volume declined in irradiated tibias (-22%; p < 0.0001) and femurs (-14%; p = 0.0586) and microarchitectural parameters were compromised. Trabecular bone volume declined in contralateral tibias (-17%; p = 0.003), and no loss was detected at the femur. Osteoclast number, apoptotic osteocyte number, and marrow adiposity were increased in irradiated bone relative to contralateral and non-irradiated bone, whereas osteoblast number was unchanged. Despite no change in osteoblast number 1 week postirradiation, dynamic bone formation indices revealed a reduction in mineralized bone surface and a concomitant increase in unmineralized osteoid surface area in irradiated bone relative to contralateral and non-irradiated control bone. Further, dose-dependent and time-dependent calvarial culture and in vitro assays confirmed that calvarial osteoblasts and osteoblast-like MC3T3 cells were relatively radioresistant, whereas calvarial osteocyte and osteocyte-like MLO-Y4 cell apoptosis was induced as early as 48 hours postirradiation (4 Gy). In osteoclastogenesis assays, radiation exposure (8 Gy) stimulated murine macrophage RAW264.7 cell differentiation, and coculture of irradiated RAW264.7 cells with MLO-Y4 or murine bone marrow cells enhanced this effect. These studies highlight the multifaceted nature of radiation-induced bone loss by demonstrating direct and systemic effects on bone and its many cell types using clinically relevant doses; they have important implications for bone health in patients treated with radiation therapy.


Assuntos
Reabsorção Óssea/patologia , Osso e Ossos/patologia , Osso e Ossos/efeitos da radiação , Membro Posterior/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Composição Corporal , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Osteoblastos/efeitos da radiação , Osteogênese/efeitos da radiação , Células RAW 264.7 , Crânio/patologia , Crânio/efeitos da radiação , Fatores de Tempo , Raios X
13.
J Orthop Res ; 33(3): 334-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25408493

RESUMO

Late-onset fragility fractures are a common complication following radiotherapy for metastatic disease and soft tissue sarcomas. Using a murine hindlimb focal irradiation model (RTx), we quantified time-dependent changes in osteoclasts and mineral apposition rate (MAR). Mice received either a single, unilateral 5 Gy exposure or four fractionated doses (4 × 5 Gy). Osteoclast numbers and MAR were evaluated histologically at 1, 2, 4, 8, 12, and 26 weeks post-RTx. Radiation induced an early, transient increase in osteoclasts followed by long-term depletion. Increased osteoclast numbers correlated temporally with trabecular resorption; the resorbed trabeculae were not later restored. Radiotherapy did not attenuate MAR at any time point. A transient, early increase in MAR was noted in both RTx groups, however, the 4 × 5 Gy group exhibited an unexpected spike in MAR eight weeks. Persistent depletion of osteoclasts permitted anabolic activity to continue unopposed, resulting in cortical thickening. These biological responses likely contribute to post-radiotherapy bone fragility via microdamage accumulation and matrix embrittlement in the absence of osteoclastic remodeling, and trabecular resorption-induced decrease in bone strength. The temporal distribution of osteoclast numbers suggests that anti-resorptive therapies may be of clinical benefit only if started prior to radiotherapy and continued through the following period of increased osteoclastic remodeling.


Assuntos
Densidade Óssea/efeitos da radiação , Fraturas Ósseas/etiologia , Osteoclastos/efeitos da radiação , Radioterapia/efeitos adversos , Animais , Feminino , Fêmur/efeitos da radiação , Membro Posterior/efeitos da radiação , Camundongos
14.
Lasers Med Sci ; 30(1): 83-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24974175

RESUMO

The increase in PGE2 production by microsomal PGE synthase-1 (mPGES-1) in CNS contributes to the severity of the inflammatory and pain responses in the model of edema formation and hyperalgesia induced by carrageenan. PGI2, alike to PGE2, plays an important role in the inflammation. Low-level laser therapy (LLLT) has been used in the treatment of inflammatory pathologies, reducing both pain and the acute inflammatory process. In this work, we studied the effect of LLLT on the expression of both mPGES-1 and IP messenger RNA (mRNA), in either subplantar or total brain tissues obtained from rats submitted to model of edema formation and hyperalgesia induced by carrageenan administration. The test sample consisted of 30 rats divided into five groups: A1 (control-saline), A2 (carrageenan-0.5 mg/paw), A3 (carrageenan-0.5 mg/paw + LLLT), A4 (carrageenan-1.0 mg/paw), and A5 (carrageenan-1.0 mg/paw + LLLT). The animals from groups A3 and A5 were irradiated 1 h after induction of inflammation by carrageenan injection. Continuous-wave red laser with wavelengths of 660 nm and dose of 7.5 J/cm(2) was used. Six hours after carrageenan-induced inflammation, mPGES-1 and prostacyclin receptor (IP) mRNA expression were significantly increased both in subplantar and brain tissues. LLLT was able to reduce both mPGES-1 and IP mRNA expression in subplantar and brain tissues. We suggest that LLLT is able to reduce both inflammation and hyperalgesia observed in the model of edema formation and hyperalgesia induced by carrageenan, by a mechanism involving the decrease in the expression of both mPGES-1 and IP.


Assuntos
Encéfalo/metabolismo , Edema/radioterapia , Membro Posterior/metabolismo , Oxirredutases Intramoleculares/genética , Lasers Semicondutores/uso terapêutico , Terapia com Luz de Baixa Intensidade , Receptores de Prostaglandina/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/efeitos da radiação , Carragenina , Regulação para Baixo , Edema/induzido quimicamente , Edema/metabolismo , Pé/patologia , Pé/efeitos da radiação , Expressão Gênica/efeitos da radiação , Membro Posterior/patologia , Membro Posterior/efeitos da radiação , Hiperalgesia/metabolismo , Hiperalgesia/radioterapia , Oxirredutases Intramoleculares/metabolismo , Masculino , Prostaglandina-E Sintases , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Prostaglandina/genética
15.
Photomed Laser Surg ; 32(11): 606-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25329504

RESUMO

OBJECTIVE: The aim of the present study was to determine whether low-level laser therapy (LLLT) at early stages postpartum could affect regeneration and degenerative processes in skeletal muscles of the dystrophic mdx mouse. BACKGROUND DATA: LLLT has been found to modulate various biological processes. It was previously shown that LLLT can markedly promote the process of skeletal muscle regeneration and angiogenesis, as well as reduce apoptosis in skeletal muscle fibers in culture. METHODS AND RESULTS: Eight newborn mdx mice were used. Ga-Al-As diode laser (810 nm) was applied at a power density of10 mW/cm(2) to the surface (area of 0.0255 cm(2)) of hindlimb muscle for 120 sec (fluence of 1.2 J/cm(2)) once a week for 4 consecutive weeks, commencing 1 week post-birth. The contralateral leg served as an untreated (sham) control. Mice were euthanized 2 days following the last laser application, and the muscles were processed for histology. Histological sections were scored for degenerative muscle foci. Statistical analysis revealed a score of 2.91±0.17 in the control, untreated group, which was significantly higher (p<0.001) than the value in the laser-treated group (1.56±0.49), indicating less degenerative foci in the laser-treated muscles. Histology also indicated regeneration (numerous myotubes) in the laser-treated mice, and no regeneration in the non-laser-treated mice. CONCLUSIONS: The results indicate that LLLT applied to mdx mice during postnatal development may have a significant beneficial effect in the induction of regenerative capacity and reduction of degenerative muscle foci in these mice, with possible direct clinical relevance.


Assuntos
Membro Posterior/efeitos da radiação , Terapia com Luz de Baixa Intensidade/métodos , Músculo Esquelético/efeitos da radiação , Regeneração/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Lasers Semicondutores , Camundongos , Camundongos Endogâmicos mdx , Neovascularização Fisiológica/efeitos da radiação
16.
PLoS One ; 9(8): e103982, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25093816

RESUMO

OBJECTIVES: Low energy shock waves have been shown to induce angiogenesis, improve left ventricular ejection fraction and decrease angina symptoms in patients suffering from chronic ischemic heart disease. Whether there is as well an effect in acute ischemia was not yet investigated. METHODS: Hind-limb ischemia was induced in 10-12 weeks old male C57/Bl6 wild-type mice by excision of the left femoral artery. Animals were randomly divided in a treatment group (SWT, 300 shock waves at 0.1 mJ/mm2, 5 Hz) and untreated controls (CTR), n = 10 per group. The treatment group received shock wave therapy immediately after surgery. RESULTS: Higher gene expression and protein levels of angiogenic factors VEGF-A and PlGF, as well as their receptors Flt-1 and KDR have been found. This resulted in significantly more vessels per high-power field in SWT compared to controls. Improvement of blood perfusion in treatment animals was confirmed by laser Doppler perfusion imaging. Receptor tyrosine kinase profiler revealed significant phosphorylation of VEGF receptor 2 as an underlying mechanism of action. The effect of VEGF signaling was abolished upon incubation with a VEGFR2 inhibitor indicating that the effect is indeed VEGFR 2 dependent. CONCLUSIONS: Low energy shock wave treatment induces angiogenesis in acute ischemia via VEGF receptor 2 stimulation and shows the same promising effects as known from chronic myocardial ischemia. It may therefore develop as an adjunct to the treatment armentarium of acute muscle ischemia in limbs and myocardium.


Assuntos
Ondas de Choque de Alta Energia/uso terapêutico , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Neovascularização Fisiológica/efeitos da radiação , Proteínas Quinases/metabolismo , Terapia por Ultrassom/métodos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Doença Aguda , Animais , Membro Posterior/efeitos da radiação , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Vasculares Periféricas/metabolismo , Doenças Vasculares Periféricas/terapia , Fosforilação
17.
Biomed Res Int ; 2013: 208912, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24350251

RESUMO

Secondary lymphedema is an intractable disease mainly caused by damage of the lymphatic system during surgery, yet studies are limited by the lack of suitable animal models. The purpose of this study was to create an improved model of secondary lymphedema in the hindlimbs of rodents with sustained effects and able to mimic human lymphedema. This was achieved by combining previously reported surgical methods and radiation to induce chronic lymphedema. Despite more radical surgical destruction of superficial and deep lymphatic vessels, surgery alone was not enough to sustain increased hindlimb volume. Radiotherapy was necessary to prolong these effects, with decreased lymphatic flow on lymphoscintigraphy, but hindlimb necrosis occurred after 4 weeks due to radiation toxicity. The applicability of this model for studies of therapeutic lymphangiogenesis was subsequently tested by injecting muscle-derived stem cells previously cocultured with the supernatant of human lymphatic endothelial cells in vitro. There was a tendency for increased lymphatic flow which significantly increased lymphatic vessel formation after cell injection, but attenuation of hindlimb volume was not observed. These results suggest that further refinement of the rodent hindlimb model is needed by titration of adequate radiation dosage, while stem cell lymphangiogenesis seems to be a promising approach.


Assuntos
Membro Posterior/patologia , Linfangiogênese/fisiologia , Vasos Linfáticos/patologia , Linfedema/patologia , Animais , Modelos Animais de Doenças , Células Endoteliais/patologia , Células Endoteliais/efeitos da radiação , Membro Posterior/efeitos da radiação , Membro Posterior/cirurgia , Humanos , Linfangiogênese/efeitos da radiação , Vasos Linfáticos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos BALB C
18.
Radiat Res ; 180(2): 205-15, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23819596

RESUMO

Radiation induced fibrosis of the skin is a late toxicity that may result in loss of function due to reduced range of motion and pain. The current study sought to determine if oral delivery of quercetin mitigates radiation-induced cutaneous injury. Female C3H/HeN mice were fed control chow or quercetin-formulated chow (1% by weight). The right hind leg was exposed to 35 Gy of X rays and the mice were followed serially to assess acute toxicity and hind leg extension. Tissue samples were collected for assessment of soluble collagen and tissue cytokines. Human and murine fibroblasts were subjected to clonogenic assays to determine the effects of quercetin on radiation response. Contractility of fibroblasts was assessed with a collagen contraction assay in the presence or absence of quercetin and transforming growth factor-ß (TGF-ß). Western blotting of proteins involved in fibroblast contractility and TGF-ß signaling were performed. Quercetin treatment significantly reduced hind limb contracture, collagen accumulation and expression of TGF-ß in irradiated skin. Quercetin had no effect on the radioresponse of fibroblasts or murine tumors, but was capable of reducing the contractility of fibroblasts in response to TGF-ß, an effect that correlated with partial stabilization of phosphorylated cofilin. Quercetin is capable of mitigating radiation induced skin fibrosis and should be further explored as a therapy for radiation fibrosis.


Assuntos
Antioxidantes/uso terapêutico , Fitoterapia , Quercetina/uso terapêutico , Protetores contra Radiação/uso terapêutico , Radiodermite/prevenção & controle , Administração Oral , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacocinética , Biotransformação , Carcinoma de Células Escamosas/radioterapia , Forma Celular , Colágeno/biossíntese , Colágeno/genética , Ensaio de Unidades Formadoras de Colônias , Avaliação Pré-Clínica de Medicamentos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Fibroblastos/efeitos da radiação , Fibrose , Genes Reporter , Membro Posterior/efeitos da radiação , Humanos , Camundongos , Camundongos Endogâmicos C3H , Células NIH 3T3/efeitos dos fármacos , Células NIH 3T3/fisiologia , Células NIH 3T3/efeitos da radiação , Quercetina/administração & dosagem , Quercetina/farmacocinética , Protetores contra Radiação/administração & dosagem , Protetores contra Radiação/farmacocinética , Radiodermite/etiologia , Radioterapia/efeitos adversos , Distribuição Aleatória , Pele/efeitos dos fármacos , Pele/patologia , Pele/efeitos da radiação , Neoplasias Cutâneas/radioterapia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/efeitos da radiação , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética
19.
Adv Exp Med Biol ; 765: 335-341, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22879053

RESUMO

Inflammatory molecules (IMs) play an important role in ionizing radiation (IR)-induced soft tissue damage. The alteration of IMs as a function of time was studied with a protein array containing 62 IMs in mouse cutaneous soft tissues exposed to 30 Gy. The results showed that: (1) 2 days after irradiation, the levels of TGF-ß1, MIP-1γ, IL-1α, and sTNF RI increased, while IGFBP-3, CXCL16, and IL-1ß decreased in IR skin as compared to control skin; (2) 21 days after IR, TGF-ß1, and MIP-1 γ, IL-1α remained high, while CXCL16 and IL-1ß remained low; (3) 3 months after IR, the cytokine pattern exhibited reversals. The levels of MIP-1γ decreased, while VCAM-1, IGFBP-3, and TGF-ß1 production increased. The data indicated that: (a) IMs change as a function of time after soft tissue irradiation; (b) changing IM levels may reflect the altered balance of the cytokine network, leading to imbalance or homeostasis; and (c) an antibody-based protein array can be used to assess multiple IMs simultaneously, making it useful for bulk screening for changes in tissue cytokine levels.


Assuntos
Membro Posterior/metabolismo , Membro Posterior/efeitos da radiação , Mediadores da Inflamação/metabolismo , Pele/metabolismo , Pele/efeitos da radiação , Lesões dos Tecidos Moles/metabolismo , Lesões dos Tecidos Moles/patologia , Animais , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Membro Posterior/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Análise Serial de Proteínas , Pele/imunologia , Fatores de Tempo
20.
Radiat Res ; 179(1): 76-88, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23216524

RESUMO

Spinal cord injury is a devastating condition with no effective treatment. The physiological processes that impede recovery include potentially detrimental immune responses and the production of reactive astrocytes. Previous work suggested that radiation treatment might be beneficial in spinal cord injury, although the method carries risk of radiation-induced damage. To overcome this obstacle we used arrays of parallel, synchrotron-generated X-ray microbeams (230 µm with 150 µm gaps between them) to irradiate an established model of rat spinal cord contusion injury. This technique is known to have a remarkable sparing effect in tissue, including the central nervous system. Injury was induced in adult female Long-Evans rats at the level of the thoracic vertebrae T9-T10 using 25 mm rod drop on an NYU Impactor. Microbeam irradiation was given to groups of 6-8 rats each, at either Day 10 (50 or 60 Gy in-beam entrance doses) or Day 14 (50, 60 or 70 Gy). The control group was comprised of two subgroups: one studied three months before the irradiation experiment (n = 9) and one at the time of the irradiations (n = 7). Hind-limb function was blindly scored with the Basso, Beattie and Bresnahan (BBB) rating scale on a nearly weekly basis. The scores for the rats irradiated at Day 14 post-injury, when using t test with 7-day data-averaging time bins, showed statistically significant improvement at 28-42 days post-injury (P < 0.038). H&E staining, tissue volume measurements and immunohistochemistry at day ≈ 110 post-injury did not reveal obvious differences between the irradiated and nonirradiated injured rats. The same microbeam irradiation of normal rats at 70 Gy in-beam entrance dose caused no behavioral deficits and no histological effects other than minor microglia activation at 110 days. Functional improvement in the 14-day irradiated group might be due to a reduction in populations of immune cells and/or reactive astrocytes, while the Day 10/Day 14 differences may indicate time-sensitive changes in these cells and their populations. With optimizations, including those of the irradiation time(s), microbeam pattern, dose, and perhaps concomitant treatments such as immunological intervention this method may ultimately reach clinical use.


Assuntos
Contusões/complicações , Membro Posterior/fisiopatologia , Membro Posterior/efeitos da radiação , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/radioterapia , Terapia por Raios X/métodos , Animais , Feminino , Método de Monte Carlo , Dosagem Radioterapêutica , Ratos , Ratos Long-Evans , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Síncrotrons , Fatores de Tempo , Terapia por Raios X/instrumentação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA