Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Radiat Res ; 195(6): 590-595, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33826738

RESUMO

Mouse models are widely used in the study of musculoskeletal radiobiology both in vivo and in vitro. Two of the most commonly used mouse strains are C57BL/6 and BALB/c. However, little is known about their equivalence in response to ionizing radiation. In this study we compare the responses of marrow stromal cells derived from both of these strains to X rays in vitro at passages 0 and 2. Colony-forming efficiency was significantly higher in BALB/c marrow stromal cells at passage 0. Radiation-induced decreases in colony-forming unit (CFU) formation at passage 0 were comparable across both strains at 0-2 Gy, but BALB/c stromal cells were more radiosensitive than C57BL/6 stromal cells at 3-7 Gy. Osteogenic differentiation at passage 2 was not affected by radiation for either strain. This work demonstrates that commonly used inbred mouse strains differ in their early-passage marrow stromal cell responses to X rays, including self-renewal and differentiation potential. This variability is an important point to consider when selecting an animal model for in vivo or in vitro study.


Assuntos
Células da Medula Óssea/citologia , Tolerância a Radiação , Células Estromais/citologia , Células Estromais/efeitos da radiação , Animais , Diferenciação Celular/efeitos da radiação , Autorrenovação Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Camundongos , Osteogênese/efeitos da radiação , Especificidade da Espécie
2.
J BUON ; 25(3): 1541-1546, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32862602

RESUMO

PURPOSE: The treatment options of endometrial hyperplasia consist of surgical, interventional and medical therapies including apoptosis-inducing agents. The purpose of the study was to evaluate the effects of ultraviolet (UV) radiation on the viability and the type of cell death on the human endometrial stromal cells (ThESC) line. METHODS: We investigated the effect of UV exposure on human endometrial stromal cell line (ThESC) on cell viability using MTT assay as well as changes in cell morphology using phase microscopy and acridine orange (AO)/ethidium bromide (EB) cell staining. RESULTS: UV treatment significantly decreased the percentage of the viable ThESC cells compared to the viability of untreated control cells using MTT assay (p<0.05). In addition, UV treatment of ThESC cells for 60 and 90 min induced high level of cell morphology disruption, followed with loss of both the cell shape and the presence of defragmented debris and stained with intense red color. CONCLUSIONS: The obtained results suggest the potential role of UV light application as additional treatment option of benign endometrium hyperplasia alone or in combination with other treatment modalities.


Assuntos
Apoptose/efeitos da radiação , Hiperplasia Endometrial/radioterapia , Células Estromais/efeitos da radiação , Morte Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Endométrio/efeitos da radiação , Feminino , Humanos , Raios Ultravioleta
3.
Bull Exp Biol Med ; 169(1): 157-161, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32488789

RESUMO

We analyzed the main properties of autologous adipose-derived stromal vascular fraction (SVF) used for the treatment of radiation-induced lesions in the rectum. No statistically significant correlation between the main characteristics of the cell product (cell number, viability) and patient's age or donor area were revealed. The stages and peculiarities of histological changes in the regenerating tissue after injection of autologous adipose tissue cells were analyzed. Morphological changes at the stages of granulation, early and complete epithelialization, and tissue maturation were described.


Assuntos
Tecido Adiposo/patologia , Lesões por Radiação , Reto/patologia , Células Estromais/patologia , Cicatrização/fisiologia , Tecido Adiposo/fisiologia , Tecido Adiposo/efeitos da radiação , Adulto , Idoso , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Mesenquimais/efeitos da radiação , Pessoa de Meia-Idade , Proctite/patologia , Proctite/fisiopatologia , Proctite/reabilitação , Lesões por Radiação/patologia , Lesões por Radiação/fisiopatologia , Lesões por Radiação/reabilitação , Radioterapia/efeitos adversos , Reepitelização/fisiologia , Reto/fisiopatologia , Células Estromais/fisiologia , Células Estromais/efeitos da radiação
4.
Arch Biochem Biophys ; 685: 108333, 2020 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-32194044

RESUMO

This study summarizes the available evidence from systematic reviews on the in vitro effects of photobiomodulation on the proliferation and differentiation of human bone and stromal cells by appraising their methodological quality. Improvements for future studies are also highlighted, with particular emphasis on in vitro protocols and cell-related characteristics. Six reviews using explicit eligibility criteria and methods selected in order to minimize bias were included. There was no compelling evidence on the cellular mechanisms of action or treatment parameters of photobiomodulation; compliance with quality assessment was poor. A rigorous description of laser parameters (wavelength, power, beam spot size, power density, energy density, repetition rate, pulse duration or duty cycle, exposure duration, frequency of treatments, and total radiant energy), exposure conditions (methods to ensure a uniform irradiation and to avoid cross-irradiation, laser-cell culture surface distance, lid presence during irradiation) and cell-related characteristics (cell type or line, isolation and culture conditions, donor-related factors where applicable, tissue source, cell phenotype, cell density, number of cell passages in culture) should be included among eligibility criteria for study inclusion. These methodological improvements will maximize the contribution of in vitro studies on the effects of photobiomodulation on human bone and stromal cells to evidence-based translational research.


Assuntos
Terapia com Luz de Baixa Intensidade , Osteócitos/metabolismo , Células Estromais/metabolismo , Animais , Diferenciação Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Humanos , Osteócitos/efeitos da radiação , Células Estromais/efeitos da radiação , Revisões Sistemáticas como Assunto
5.
Sci Rep ; 9(1): 14134, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31575959

RESUMO

Radiation-induced intestinal injury (RIII) constitutes a crucial clinical element of acute radiation syndrome with life-threatening implications posing challenges in devising effective medical countermeasures. Herein, we report the potential of 7, 8-diacetoxy-4-methylthiocoumarin (DAMTC) to mitigate RIII following total-body irradiation (TBI) in C57BL/6 mice and underlying mechanisms. Administration of DAMTC 24 hours post TBI facilitated structural reconstitution and restoration of functional absorption linked to alleviation of radiation-induced apoptotic death of intestinal crypt progenitor/stem (ICPS) and villus stromal cells through induction of Bcl-2 family-mediated anti-apoptotic signalling. Reduction in TBI-induced DNA damage accumulation coupled with inhibition of cell cycle arrest through stimulation of anti-p53- and anti-p21-dependent synergistic signalling protected ICPS cells from radiation injury. Enhanced proliferation of crypt stem cells, induction of anti-oxidant defence, subjugation of TBI-induced lipid peroxidation and phenotypic polarization of intestinal macrophages to anti-inflammatory M2 class underlie amelioration of RIII. Stimulation of multiple mitigative signalling processes by DAMTC appeared to be associated with enhanced protein acetylation, an important regulator of cellular responses to radiation damage. Our findings establish the mitigative potential of DAMTC against RIII by hyper-acetylation-mediated epigenetic regulation, which triggers axes of anti-apoptotic and pro-survival pathways, enabling proliferation and maintenance of ICPS cells leading to epithelial regeneration.


Assuntos
Anormalidades Induzidas por Radiação/tratamento farmacológico , Síndrome Aguda da Radiação/tratamento farmacológico , Cumarínicos/farmacologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos da radiação , Anormalidades Induzidas por Radiação/metabolismo , Síndrome Aguda da Radiação/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Feminino , Trato Gastrointestinal/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/efeitos da radiação , Irradiação Corporal Total/efeitos adversos
6.
Sci Rep ; 9(1): 12094, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31431660

RESUMO

Decidualization is characterized by the differentiation of endometrial stromal cells (eSCs), which is critical for embryo implantation and maintenance of pregnancy. In the present study, we investigated the possible effect of simulated microgravity (SM) on the process of proliferation and in vitro decidualization using primary human eSCs. Exposure to SM for 36 h decreased the proliferation and migration of eSCs significantly, without inducing cell death and changes in cell cycle progression. The phosphorylation of Akt decreased under SM conditions in human eSCs, accompanied by a simultaneous decrease in the level of matrix metalloproteinase (MMP)-2 and FOXO3a. Treatment with Akti, an Akt inhibitor, decreased MMP-2 expression, but not FOXO3a expression. The decreased level of FOXO3a under SM conditions impeded autophagic flux by reducing the levels of autophagy-related genes. In addition, pre-exposure of eSCs to SM significantly inhibited 8-Br-cAMP induced decidualization, whereas restoration of the growth status under SM conditions by removing 8-Br-cAMP remained unchanged. Treatment of human eSCs with SC-79, an Akt activator, restored the reduced migration of eSCs and decidualization under SM conditions. In conclusion, exposure to SM inhibited decidualization in eSCs by decreasing proliferation and migration through Akt/MMP and FOXO3a/autophagic flux.


Assuntos
Autofagia/genética , Endométrio/crescimento & desenvolvimento , Proteína Forkhead Box O3/genética , Metaloproteinase 2 da Matriz/genética , Proteína Oncogênica v-akt/genética , Adulto , Autofagia/efeitos da radiação , Diferenciação Celular/genética , Diferenciação Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Células Cultivadas , Decídua/crescimento & desenvolvimento , Decídua/metabolismo , Decídua/efeitos da radiação , Implantação do Embrião/genética , Implantação do Embrião/efeitos da radiação , Endométrio/metabolismo , Endométrio/efeitos da radiação , Meio Ambiente , Feminino , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Humanos , Pessoa de Meia-Idade , Fosforilação/efeitos da radiação , Gravidez , Células Estromais/metabolismo , Células Estromais/efeitos da radiação , Simulação de Ausência de Peso
7.
Semin Oncol ; 46(3): 254-260, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31383368

RESUMO

The broad use of radiotherapy (RT) in the management of solid human tumors is based on its ability to damage cellular macromolecules, particularly the DNA, effectively inducing growth arrest and cell death locally in irradiated tumor cells. However, bystander effects, such as the transmission of lethal signals between cells via gap junctions or the production of diffusible cytotoxic mediators, can also contribute to the local antineoplastic action of RT. Traditionally, RT has been considered to exert immunosuppressive effects on the host. This idea largely stems from the radiosensitivity of quiescent lymphocytes and on the use of total body irradiation as part of myeloablative conditioning regimens preceding hematopoietic stem cell transplantation. Additionally, the occurrence of the so-called "abscopal effect," where nonirradiated distant lesions display effects of RT response, suggests that RT may also induce tumor immunization. Several RT-induced effects on cancer, immune and stromal cells, contribute to the abscopal effect: (1) induction of "immunogenic cell death", with release of tumor-associated antigens, (2) alterations of cancer cell immunophenotype, and (3) modulation of the tumor microenvironment. Damage and death of cancer cells leads to the surface exposure of immunogenic molecules as well as the release of damage associated molecular patterns such as adenosine triphosphate or High-Mobility-Group-Protein B1, and potentially tumor antigens that activate the innate and adaptive immune systems. Moreover, nuclear release and cytoplasmic sensing of altered nucleic acids via cyclic GMP-AMP Synthase/Stimulator of Interferon Genes is connected to the secretion of cytokines that support innate and adaptive antitumor immunity. As a result of the above, irradiated tumor cells may potentially act as an "in situ vaccine."


Assuntos
Morte Celular/efeitos da radiação , Neoplasias/radioterapia , Tolerância a Radiação/imunologia , Células Estromais/efeitos da radiação , Antígenos de Neoplasias/imunologia , Morte Celular/imunologia , Proteína HMGB1/genética , Proteína HMGB1/imunologia , Humanos , Imunofenotipagem , Terapia de Imunossupressão , Linfócitos/imunologia , Linfócitos/efeitos da radiação , Neoplasias/imunologia , Células Estromais/imunologia , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação , Irradiação Corporal Total/efeitos adversos
8.
Sci Adv ; 5(3): eaav1281, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30906864

RESUMO

Corneal injuries are common causes of visual impairment worldwide. Accordingly, there is an unmet need for transparent biomaterials that have high adhesion, cohesion, and regenerative properties. Herein, we engineer a highly biocompatible and transparent bioadhesive for corneal reconstruction using a visible light cross-linkable, naturally derived polymer, GelCORE (gel for corneal regeneration). The physical properties of GelCORE could be finely tuned by changing prepolymer concentration and photocrosslinking time. GelCORE revealed higher tissue adhesion compared to commercial adhesives. Furthermore, in situ photopolymerization of GelCORE facilitated easy delivery to the cornea, allowing for bioadhesive curing precisely according to the required geometry of the defect. In vivo experiments, using a rabbit stromal defect model, showed that bioadhesive could effectively seal corneal defects and induce stromal regeneration and re-epithelialization. Overall, GelCORE has many advantages including low cost and ease of production and use. This makes GelCORE a promising bioadhesive for corneal repair.


Assuntos
Lesões da Córnea/cirurgia , Hidrogéis/farmacologia , Regeneração/efeitos dos fármacos , Procedimentos Cirúrgicos sem Sutura/métodos , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Córnea/efeitos dos fármacos , Córnea/patologia , Córnea/cirurgia , Lesões da Córnea/patologia , Humanos , Hidrogéis/efeitos da radiação , Luz , Coelhos , Regeneração/efeitos da radiação , Células Estromais/efeitos da radiação
9.
Front Immunol ; 10: 193, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30828330

RESUMO

In recent decades, there has been substantial growth in our understanding of the immune system and its role in tumor growth and overall survival. A central finding has been the cross-talk between tumor cells and the surrounding environment or stroma. This tumor stroma, comprised of various cells, and extracellular matrix (ECM), has been shown to aid in suppressing host immune responses against tumor cells. Through immunosuppressive cytokine secretion, metabolic alterations, and other mechanisms, the tumor stroma provides a complex network of safeguards for tumor proliferation. With recent advances in more effective, localized treatment, radiation therapy (XRT) has allowed for strategies that can effectively alter and ablate tumor stromal tissue. This includes promoting immunogenic cell death through tumor antigen release to increasing immune cell trafficking, XRT has a unique advantage against the tumoral immune evasion mechanisms that are orchestrated by stromal cells. Current studies are underway to elucidate pathways within the tumor stroma as potential targets for immunotherapy and chemoradiation. This review summarizes the effects of tumor stroma in tumor immune evasion, explains how XRT may help overcome these effects, with potential combinatorial approaches for future treatment modalities.


Assuntos
Neoplasias/patologia , Neoplasias/radioterapia , Células Estromais/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Animais , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/efeitos da radiação , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Matriz Extracelular/efeitos da radiação , Humanos , Imunidade , Imunomodulação/efeitos da radiação , Neoplasias/imunologia , Tolerância a Radiação/imunologia , Tolerância a Radiação/efeitos da radiação , Radioterapia , Células Estromais/imunologia , Microambiente Tumoral/imunologia
10.
Clin Cancer Res ; 25(10): 2969-2974, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30723144

RESUMO

The development of cancers and their response to radiation are intricately linked to the tumor microenvironment (TME) in which they reside. Tumor cells, immune cells, and stromal cells interact with each other and are influenced by the microbiome and metabolic state of the host, and these interactions are constantly evolving. Stromal cells not only secrete extracellular matrix and participate in wound contraction, but they also secrete fibroblast growth factors (FGF), which mediate macrophage differentiation. Tumor-associated macrophages migrate to hypoxic areas and secrete vascular endothelial growth factor (VEGF) to promote angiogenesis. The microbiome and its byproducts alter the metabolic milieu by shifting the balance between glucose utilization and fatty acid oxidation, and these changes subsequently influence the immune response in the TME. Not only does radiation exert cell-autonomous effects on tumor cells, but it influences both the tumor-promoting and tumor-suppressive components in the TME. To gain a deeper understanding of how the TME influences the response to radiation, the American Society for Radiation Oncology and the American Association of Cancer Research organized a scientific workshop on July 26-27, 2018, to discuss how the microbiome, the immune response, the metabolome, and the stroma all shift the balance between radiosensitivity and radioresistance. The proceedings from this workshop are discussed here and highlight recent discoveries in the field, as well as the most important areas for future research.


Assuntos
Neoplasias/patologia , Neoplasias/radioterapia , Radioterapia (Especialidade) , Microambiente Tumoral/efeitos da radiação , Gerenciamento Clínico , Humanos , Imunomodulação/efeitos da radiação , Neoplasias/imunologia , Radioterapia (Especialidade)/métodos , Fatores de Risco , Células Estromais/imunologia , Células Estromais/metabolismo , Células Estromais/efeitos da radiação , Resultado do Tratamento , Microambiente Tumoral/imunologia
11.
J Toxicol Environ Health A ; 82(3): 157-162, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30663538

RESUMO

Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is a primary transcription factor which plays a key role in several cellular processes including proliferation and survival. It is well-known that exposure to non-ionizing radiofrequency fields (RF), which are ubiquitous, interact with cellular components. The aim of the study was thus to examine whether exposure of mouse bone marrow stromal cells (BMSC) to RF also resulted in cellular interactions. BMSC were exposed to 900 MHz RF at 120 µW/cm2 power intensity for 4 hr/day for 5 consecutive days. The relative protein expression levels of NF-κB in the cytoplasm and nucleus of RF-exposed cells were compared to non-RF-exposed controls. At 30 min post-RF exposure a significant decrease in protein expression of NF-κB in the cytoplasm was accompanied by a concomitant increase in nuclear NF-κB protein expression levels. Similar responses were noted in the cytoplasm and nuclear NF-κB levels at 2 hr with a return to control concentrations in primary transcription factor at 24 hr post-RF treatment. Daily incubation of BAY 11-7082 an inhibitor of NF-κB for 90 min for 5 days followed by RF each day prevented the fall in cytoplasmic NF-κB and rise in nuclear primary transcription factor at 30 min and 2 hr. There were no marked alterations at 24 hr. Data showed that the effects of RF treatment on BMSC involved transient activation of NF-κB which may be attributed to RF-mediated cellular perturbation as evidenced by consequences of BAY 11-7082 inhibition.


Assuntos
Células da Medula Óssea/efeitos da radiação , Células Cultivadas/efeitos da radiação , NF-kappa B/efeitos da radiação , Ondas de Rádio/efeitos adversos , Transdução de Sinais , Células Estromais/efeitos da radiação , Animais , Camundongos
12.
Nat Commun ; 9(1): 2783, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30018330

RESUMO

The importance of the tumor-associated stroma in cancer progression is clear. However, it remains uncertain whether early events in the stroma are capable of initiating breast tumorigenesis. Here, we show that in the mammary glands of non-tumor bearing mice, stromal-specific phosphatase and tensin homolog (Pten) deletion invokes radiation-induced genomic instability in neighboring epithelium. In these animals, a single dose of whole-body radiation causes focal mammary lobuloalveolar hyperplasia through paracrine epidermal growth factor receptor (EGFR) activation, and EGFR inhibition abrogates these cellular changes. By analyzing human tissue, we discover that stromal PTEN is lost in a subset of normal breast samples obtained from reduction mammoplasty, and is predictive of recurrence in breast cancer patients. Combined, these data indicate that diagnostic or therapeutic chest radiation may predispose patients with decreased stromal PTEN expression to secondary breast cancer, and that prophylactic EGFR inhibition may reduce this risk.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Mamárias Experimentais/genética , PTEN Fosfo-Hidrolase/genética , Tolerância a Radiação/genética , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/radioterapia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Transformação Celular Neoplásica , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Raios gama/efeitos adversos , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/efeitos da radiação , Humanos , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/efeitos da radiação , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/efeitos da radiação , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/radioterapia , Camundongos , PTEN Fosfo-Hidrolase/deficiência , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/efeitos da radiação
13.
Eur Rev Med Pharmacol Sci ; 22(9): 2860-2868, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29771444

RESUMO

OBJECTIVE: The relationship between adiponectin (APN) pathway and Wnt pathway was explored through BMSCs, and the effect of low-level laser irradiation (LLLI) on bone marrow stromal cells (BMSCs) and its mechanism were further studied. MATERIALS AND METHODS: 3-week-old Sprague-Dawley (SD) rats were selected, and mesenchymal stem cells were separately cultured and purified. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to analyze cell proliferation. After osteogenic and adipogenic induction, cultures were conducted, respectively, cells were stained with alizarin red and oil red O. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect the expressions of osteogenesis-related genes, runt-related transcription factor 2 (RUNX2), and osteocalcin (OC) and those of adipogenesis-related genes, peroxisome proliferator-activated receptor-gamma (PPARγ) and CCAAT/enhancer-binding protein alpha (c/EBPα). Western blotting was used to detect the expressions of ß-catenin in the cytoplasm and nucleus. The lentiviral expression vector of adiponectin receptors (APN-R) was constructed, and the expression of APN receptor genes was silenced. The expressions of ß-catenin in APN receptors and the nucleus within cells were detected. RESULTS: LLLI promoted the bone formation by inducing the differentiation direction of mesenchymal stem cells, increasing the number of osteoblasts in the bone marrow and inhibiting the reduction of the number of adipocytes. LLLI regulates the Wnt pathway, promotes the entry of ß-catenin into the nucleus, activates the osteogenic effect of the Wnt pathway so as to promote the bone formation of osteoblasts and inhibit bone resorption of osteoclasts. LLLI promotes the entry of ß-catenin into the nucleus and the osteogenic differentiation of BMSCs through the APN pathway. CONCLUSIONS: In summary, LLLI can promote osteogenesis and inhibit adipocytes formation, thus attenuating bone resorption of osteoclasts. The mechanism of LLLI is that it promotes the entry of ß-catenin into the nucleus and regulates the Wnt pathway and the differentiation direction of mesenchymal stem cells through the APN signal pathway, thus promoting bone formation.


Assuntos
Adiponectina/metabolismo , Células da Medula Óssea/metabolismo , Terapia com Luz de Baixa Intensidade/métodos , Osteoblastos/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Células da Medula Óssea/efeitos da radiação , Diferenciação Celular/fisiologia , Diferenciação Celular/efeitos da radiação , Células Cultivadas , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/efeitos da radiação , Osteoblastos/efeitos da radiação , Osteogênese/fisiologia , Osteogênese/efeitos da radiação , Ratos , Ratos Sprague-Dawley , Células Estromais/metabolismo , Células Estromais/efeitos da radiação , Via de Sinalização Wnt/efeitos da radiação , beta Catenina/efeitos da radiação
14.
Reproduction ; 155(6): 553-562, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29636407

RESUMO

Radiation damage due to total body irradiation (TBI) or targeted abdominal radiation can deplete ovarian follicles and accelerate reproductive aging. We characterized a mouse model of low-dose TBI to investigate how radiation affects the follicular and stromal compartments of the ovary. A single TBI dose of either 0.1 Gy or 1 Gy (Cesium-137 γ) was delivered to reproductively adult CD1 female mice, and sham-treated mice served as controls. Mice were euthanized either 2 weeks or 5 weeks post exposure, and ovarian tissue was harvested. To assess the ovarian reserve, we classified and counted the number of morphologically normal follicles in ovarian histologic sections for all experimental cohorts using an objective method based on immunohistochemistry for an oocyte-specific protein (MSY2). 0.1 Gy did not affect that total number of ovarian follicles, whereas 1 Gy resulted in a dramatic loss. At two weeks, there was a significant reduction in all preantral follicles, but early antral and antral follicles were still present. By five weeks, there was complete depletion of all follicle classes. We examined stromal quality using histologic stains to visualize ovarian architecture and fibrosis and by immunohistochemistry and quantitative microscopy to assess cell proliferation, cell death and vasculature. There were no differences in the ovarian stroma across cohorts with respect to these markers, indicating that this compartment is more radio-resistant relative to the germ cells. These findings have implications for reproductive health and the field of fertility preservation because the radiation doses we examined mimic scatter doses experienced in typical therapeutic regimens.


Assuntos
Folículo Ovariano/patologia , Células Estromais/citologia , Irradiação Corporal Total , Animais , Células Cultivadas , Feminino , Camundongos , Folículo Ovariano/efeitos da radiação , Células Estromais/efeitos da radiação
15.
Lasers Med Sci ; 33(6): 1215-1223, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29623521

RESUMO

It is well established that laser phototherapy (LP) is contraindicated directly over cancer cells, due to its bio modulatory effects in cell and blood vessel proliferation. The aim of the present study was to analyze the influence of typical low-level laser therapy (LLLT) and high intensity laser therapy (HILT) and an in-between dose of 9 J on collagen fibers and blood vessels content in melanoma tumors (B16F10) implanted in mice. Melanoma tumor cells were injected in male Balb C mice which were distributed in four groups: control (no irradiated) or irradiated by 3, 9, or 21 J (150; 450, or 1050 J/cm2). LP was performed in daily sessions for 3 days with a InGaAlP-660 nm (mean output: 50 mW, spot size: 2 mm2). Tumor volume was analyzed using (1) picrosirius staining to quantify collagen fibers content and (2) Verhoeff's method to quantify blood vessels content. Tumor growth outcome measured in the 3-J group was not significantly different from controls. Nine and 21-J groups, presented significant and dose-dependent increases in tumor volume. Quantitative analysis of the intensity of collagen fibers and their organization in stroma and peri-tumoral microenvironment showed significant differences between irradiated and control group. Blood vessels count of 21-J group outnumbered the other groups. High doses (≥ 9 J) of LP showed a dose-dependent tumor growth, different collagen fibers characteristics, and eventually blood vessel growth, while a typical LLLT dose (3 J) appeared harmless on melanoma cell activity.


Assuntos
Tecido Conjuntivo/patologia , Tecido Conjuntivo/efeitos da radiação , Terapia com Luz de Baixa Intensidade/métodos , Melanoma Experimental/patologia , Animais , Proliferação de Células/efeitos da radiação , Colágeno Tipo I/metabolismo , Relação Dose-Resposta à Radiação , Colágenos Fibrilares/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Coloração e Rotulagem , Células Estromais/patologia , Células Estromais/efeitos da radiação , Carga Tumoral/efeitos da radiação
16.
J Tissue Eng Regen Med ; 12(6): 1352-1362, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29603903

RESUMO

A highly interesting source for adult stem cells is adipose tissue, from which the stromal vascular fraction (SVF)-a heterogeneous cell population including the adipose-derived stromal/stem cells-can be obtained. To enhance the regenerative potential of freshly isolated SVF cells, low-level light therapy (LLLT) was used. The effects of pulsed blue (475 nm), green (516 nm), and red (635 nm) light from light-emitting diodes applied on freshly isolated SVF were analysed regarding cell phenotype, cell number, viability, adenosine triphosphate content, cytotoxicity, and proliferation but also osteogenic, adipogenic, and proangiogenic differentiation potential. The colony-forming unit fibroblast assay revealed a significantly increased colony size after LLLT with red light compared with untreated cells, whereas the frequency of colony-forming cells was not affected. LLLT with green and red light resulted in a stronger capacity to form vascular tubes by SVF when cultured within 3D fibrin matrices compared with untreated cells, which was corroborated by increased number and length of the single tubes and a significantly higher concentration of vascular endothelial growth factor. Our study showed beneficial effects after LLLT on the vascularization potential and proliferation capacity of SVF cells. Therefore, LLLT using pulsed light-emitting diode light might represent a new approach for activation of freshly isolated SVF cells for direct clinical application.


Assuntos
Tecido Adiposo/citologia , Separação Celular , Terapia com Luz de Baixa Intensidade , Diferenciação Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Feminino , Humanos , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos da radiação , Células Estromais/citologia , Células Estromais/efeitos da radiação
17.
Stem Cells Dev ; 27(18): 1268-1277, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29609526

RESUMO

Elucidation of the spaceflight (SF) effects on the adult stem and progenitor cells is an important goal in space biology and medicine. A unique opportunity for this was provided by project "BION-M1". The purpose of this study was to evaluate the effects of 30-day SF on biosatellite, 7-day recovery (SFR), and subsequent ground control (GC) experiment on the mononuclear cells (MNCs) from C57/BI/6N murine tibia bone marrow. Also, hematopoietic and stromal precursor functions were characterized ex vivo. There was no significant difference in the total MNC number between experimental groups. After SF, immunophenotyping revealed an increase of large-sized CD45+MNCs corresponded to committed hematopoietic progenitors. The total hematopoietic colony-forming unit (CFU) number decreased after SF and did not restore after 7 day of recovery due to predominant reduction of bi- and multipotent CFUs and primitive burst-forming units in favor of unipotent CFUs. Functional activity of stromal precursors in vitro was only slightly altered. SF cells displayed the enhanced expression of alkaline phosphatase. The data of the GC experiment demonstrated the preservation of the functional activity of progenitor cells from mice bone marrow. The activation of erythropoiesis in expense of burst-forming units of erythrocytes elevation was detected. After 7 days of recovery, the number of colony-forming units of fibroblast (CFUs-f) was similar to the vivarium control, while the proliferative activity of bone marrow stromal precursors decreased. The present study demonstrated that certain hematopoietic progenitors are susceptible to SF factors, while the stromal precursors displayed a certain degree of resistance. These data indicate mild and reversible alterations of bone marrow progenitors after SF.


Assuntos
Células da Medula Óssea/efeitos da radiação , Células-Tronco Hematopoéticas/efeitos da radiação , Voo Espacial , Células Estromais/efeitos da radiação , Animais , Ensaio de Unidades Formadoras de Colônias , Eritropoese/efeitos da radiação , Fibroblastos/efeitos da radiação , Imunofenotipagem , Camundongos
18.
Cancer Immunol Res ; 6(4): 422-433, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29472271

RESUMO

The cGAS-STING cytosolic DNA sensing pathway may play an integral role in the initiation of antitumor immune responses. Studies evaluating the immunogenicity of various cyclic dinucleotide (CDN) STING agonists administered by intratumoral (i.t.) injection showed potent induction of inflammation, tumor necrosis, and, in some cases, durable tumor-specific adaptive immunity. However, the specific immune mechanisms underlying these responses remain incompletely defined. The majority of these studies have focused on the effect of CDNs on immune cells but have not conclusively interrogated the role of stromal cells in the acute rejection of the CDN-injected tumor. Here, we revealed a mechanism of STING agonist-mediated tumor response that relied on both stromal and immune cells to achieve tumor regression and clearance. Using knockout and bone marrow chimeric mice, we showed that although bone marrow-derived TNFα was necessary for CDN-induced necrosis, STING signaling in radioresistant stromal cells was also essential for CDN-mediated tumor rejection. These results provide evidence for crosstalk between stromal and hematopoietic cells during CDN-mediated tumor collapse after i.t. administration. These mechanistic insights may prove critical in the clinical development of STING agonists. Cancer Immunol Res; 6(4); 422-33. ©2018 AACR.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Membrana/agonistas , Neoplasias/etiologia , Neoplasias/metabolismo , Nucleotídeos Cíclicos/farmacologia , Tolerância a Radiação , Células Estromais/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Medula Óssea/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Inata , Interferon beta/metabolismo , Melanoma Experimental , Camundongos , Camundongos Knockout , Necrose/metabolismo , Necrose/patologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Tolerância a Radiação/efeitos dos fármacos , Tolerância a Radiação/genética , Transdução de Sinais/efeitos dos fármacos , Células Estromais/patologia , Células Estromais/efeitos da radiação , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
19.
Artigo em Inglês | MEDLINE | ID: mdl-28676262

RESUMO

This study examined whether non-ionizing radiofrequency fields (RF) exposure is capable of inducing poly (ADP-ribose) polymerase-1 (PARP-1) in bone marrow stromal cells (BMSCs) and whether it plays a role in RF-induced adaptive response (AR). Bone marrow stromal cells (BMSCs) were exposed to 900MHz RF at 120µW/cm2 power flux density for 3h/day for 5days and then challenged with a genotoxic dose of 1.5Gy gamma-radiation (GR). Some cells were also treated with 3-aminobenzamide (3-AB, 2mM final concentration), a potent inhibitor of PARP-1. Un-exposed and sham (SH)-exposed control cells as well as positive control cells exposed to gamma radiation (GR) were included in the experiments. The expression of PARP-1 mRNA and its protein levels as well as single strand breaks in the DNA and the kinetics of their repair were evaluated at several times after exposures. The results indicated the following. (a) Cells exposed to RF alone showed significantly increased PARP-1 mRNA expression and its protein levels compared with those exposed to SH- and GR alone. (b) Treatment of RF-exposed cells with 3-AB had diminished such increase in PARP-1. (c) Cells exposed to RF+GR showed significantly decreased genetic damage as well as faster kinetics of repair compared with those exposed to GR alone. (d) Cells exposed to RF+3-AB+GR showed no such decrease in genetic damage. Thus, the overall date suggested that non-ionizing RF exposure was capable of inducing PARP-1 which has a role in RF-induced AR.


Assuntos
Adaptação Fisiológica/efeitos da radiação , Células da Medula Óssea/efeitos da radiação , Quebras de DNA de Cadeia Simples , Reparo do DNA , Poli(ADP-Ribose) Polimerase-1/biossíntese , Ondas de Rádio/efeitos adversos , Células Estromais/efeitos da radiação , Adaptação Fisiológica/genética , Animais , Células da Medula Óssea/patologia , Células Cultivadas , Ensaio Cometa , Indução Enzimática , Raios gama/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos , Poli(ADP-Ribose) Polimerase-1/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Estromais/patologia
20.
Clin Cancer Res ; 23(19): 5948-5958, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28679765

RESUMO

Purpose: Evidence suggests that PD-L1 can be induced with radiotherapy and may be an immune escape mechanism in cancer. Monitoring this response is limited, as repetitive biopsies during therapy are impractical, dangerous, and miss tumor stromal cells. Monitoring PD-L1 expression in both circulating tumor cells (CTCs) and circulating stromal cells (CStCs) in blood-based biopsies might be a practical alternative for sequential, noninvasive assessment of changes in tumor and stromal cells.Experimental Design: Peripheral blood was collected before and after radiotherapy from 41 patients with lung cancer, as were primary biopsies. We evaluated the expression of PD-L1 and formation of RAD50 foci in CTCs and a CStC subtype, cancer-associated macrophage-like cells (CAMLs), in response to DNA damage caused by radiotherapy at the tumor site.Results: Only 24% of primary biopsies had sufficient tissue for PD-L1 testing, tested with IHC clones 22c3 and 28-8. A CTC or CAML was detectable in 93% and 100% of samples, prior to and after radiotherapy, respectively. RAD50 foci significantly increased in CTCs (>7×, P < 0.001) and CAMLs (>10×, P = 0.001) after radiotherapy, confirming their origin from the radiated site. PD-L1 expression increased overall, 1.6× in CTCs (P = 0.021) and 1.8× in CAMLs (P = 0.004): however, individual patient PD-L1 expression varied, consistently low/negative (51%), consistently high (17%), or induced (31%).Conclusions: These data suggest that RAD50 foci formation in CTCs and CAMLs may be used to track cells subjected to radiation occurring at primary tumors, and following PD-L1 expression in circulating cells may be used as a surrogate for tracking adaptive changes in immunotherapeutic targets. Clin Cancer Res; 23(19); 5948-58. ©2017 AACR.


Assuntos
Antígeno B7-H1/sangue , Enzimas Reparadoras do DNA/sangue , Proteínas de Ligação a DNA/sangue , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/radioterapia , Hidrolases Anidrido Ácido , Adulto , Idoso , Biomarcadores Tumorais/sangue , Biópsia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Células Neoplásicas Circulantes/efeitos da radiação , Células Estromais/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA