Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3018, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38589357

RESUMO

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced acute GI syndrome. Through single-cell RNA-sequencing of the irradiated mouse small intestine, we find that p53 target genes are specifically enriched in regenerating epithelial cells that undergo fetal-like reversion, including revival stem cells (revSCs) that promote animal survival after severe damage of the GI tract. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce fetal-like revSCs. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells and is controlled by an Mdm2-mediated negative feedback loop. Together, our findings reveal that p53 suppresses severe radiation-induced GI injury by promoting fetal-like reprogramming of irradiated intestinal epithelial cells.


Assuntos
Lesões por Radiação , Proteína Supressora de Tumor p53 , Camundongos , Animais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Intestinos , Trato Gastrointestinal/metabolismo , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Células-Tronco/metabolismo , Apoptose/genética
2.
Int J Radiat Oncol Biol Phys ; 118(5): 1315-1327, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38104870

RESUMO

PURPOSE: Despite aggressive multimodal treatment that typically includes definitive or adjuvant radiation therapy (RT), locoregional recurrence rates approach 50% for patients with locally advanced human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC). Thus, more effective therapeutics are needed to improve patient outcomes. We evaluated the radiosensitizing effects of ataxia telangiectasia and RAD3-related (ATR) inhibitor (ATRi) BAY 1895344 in preclinical models of HNSCC. METHODS AND MATERIALS: Murine and human HPV-negative HNSCC cells (MOC2, MOC1, JHU-012) were treated with vehicle or ATRi with or without 4 Gy. Checkpoint kinase 1 phosphorylation and DNA damage (γH2AX) were evaluated by Western blot, and ATRi half-maximal inhibitory concentration was determined by MTT assay for HNSCC cells and immortalized murine oral keratinocytes. In vitro radiosensitization was tested by clonogenic assay. Cell cycle distribution and mitotic catastrophe were evaluated by flow cytometry. Mitotic aberrations were quantified by fluorescent microscopy. Tumor growth delay and survival were assessed in mice bearing MOC2 or JHU-012 transplant tumors treated with vehicle, ATRi, RT (10 Gy × 1 or 8 Gy × 3), or combined ATRi + RT. RESULTS: ATRi caused dose-dependent reduction in checkpoint kinase 1 phosphorylation at 1 hour post-RT (4 Gy) and dose-dependent increase in γH2AX at 18 hours post-RT. Addition of RT to ATRi led to decreased BAY 1895344 half-maximal inhibitory concentration in HNSCC cell lines but not in normal tissue surrogate immortalized murine oral keratinocytes. Clonogenic assays demonstrated radiosensitization in the HNSCC cell lines. ATRi abrogated the RT-induced G2/M checkpoint, leading to mitosis with unrepaired DNA damage and increased mitotic aberrations (multinucleated cells, micronuclei, nuclear buds, nucleoplasmic bridges). ATRi and RT significantly delayed tumor growth in MOC2 and JHU-012 in vivo models, with improved overall survival in the MOC2 model. CONCLUSIONS: These findings demonstrated that BAY 1895344 increased in vitro and in vivo radiosensitivity in HPV-negative HNSCC preclinical models, suggesting therapeutic potential warranting evaluation in clinical trials for patients with locally advanced or recurrent HPV-negative HNSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Morfolinas , Infecções por Papillomavirus , Pirazóis , Radiossensibilizantes , Humanos , Animais , Camundongos , Carcinoma de Células Escamosas de Cabeça e Pescoço/radioterapia , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Quinase 1 do Ponto de Checagem/metabolismo , Recidiva Local de Neoplasia/tratamento farmacológico , Radiossensibilizantes/farmacologia , Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/radioterapia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo
3.
Cancer Res Commun ; 3(12): 2455-2467, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-37982576

RESUMO

Approximately half of patients with cancer receive radiotherapy and, as cancer survivorship increases, the low rate of radiation-associated sarcomas is rising. Pharmacologic inhibition of p53 has been proposed as an approach to ameliorate acute injury of normal tissues from genotoxic therapies, but how this might impact the risk of therapy-induced cancer and normal tissue injuries remains unclear. We utilized mice that express a doxycycline (dox)-inducible p53 short hairpin RNA to reduce Trp53 expression temporarily during irradiation. Mice were placed on a dox diet 10 days prior to receiving 30 or 40 Gy hind limb irradiation in a single fraction and then returned to normal chow. Mice were examined weekly for sarcoma development and scored for radiation-induced normal tissue injuries. Radiation-induced sarcomas were subjected to RNA sequencing. Following single high-dose irradiation, 21% of animals with temporary p53 knockdown during irradiation developed a sarcoma in the radiation field compared with 2% of control animals. Following high-dose irradiation, p53 knockdown preserves muscle stem cells, and increases sarcoma development. Mice with severe acute radiation-induced injuries exhibit an increased risk of developing late persistent wounds, which were associated with sarcomagenesis. RNA sequencing revealed radiation-induced sarcomas upregulate genes related to translation, epithelial-mesenchymal transition (EMT), inflammation, and the cell cycle. Comparison of the transcriptomes of human and mouse sarcomas that arose in irradiated tissues revealed regulation of common gene programs, including elevated EMT pathway gene expression. These results suggest that blocking p53 during radiotherapy could minimize acute toxicity while exacerbating late effects including second cancers. SIGNIFICANCE: Strategies to prevent or mitigate acute radiation toxicities include pharmacologic inhibition of p53 and other cell death pathways. Our data show that temporarily reducing p53 during irradiation increases late effects including sarcomagenesis.


Assuntos
Lesões por Radiação , Sarcoma , Humanos , Camundongos , Animais , Proteína Supressora de Tumor p53/genética , Sarcoma/genética , Ciclo Celular , Dano ao DNA
4.
Methods Cell Biol ; 180: 147-175, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37890927

RESUMO

Given the potential risk of radiological terrorism and disasters, it is essential to develop plans to prepare for such events. In these hazardous scenarios, radiation-induced gastrointestinal (GI) syndrome is one of the many manifestations that may happen after the organism is exposed to a lethal dose of ionizing radiation. Therefore, it is critical to better understand how the intestinal tissues initiate and orchestrate regeneration following severe radiation injury. In this chapter, we aimed to provide several key considerations for researchers who utilize histological assessment to study radiation-induced intestinal injury. Rigor and reproducibility are critical in experimental design and can be achieved by maintaining proper radiation administration, maintaining consistency in sample collection, and selecting and using appropriate controls. We also provided technical details of histological preparation of the intestines with tips on dissecting, cleaning, fixing, and preserving. Step-by-step descriptions of both bundling and Swiss rolling are provided with discussion on how to choose between the two approaches. In the following section, we detailed several histological assessment methods and then provided suggestions on how to use histological assessment to study cellular dynamics in the small intestines. Finally, we touched on some non-histological assessments. We hope that the information provided in this chapter will contribute to the research society of radiation-induced intestinal injury with an ultimate goal of promoting the development of radiation countermeasures against the GI acute radiation syndrome.


Assuntos
Intestino Delgado , Intestinos , Reprodutibilidade dos Testes , Intestinos/patologia , Radiação Ionizante
5.
bioRxiv ; 2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37162959

RESUMO

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced GI injury. Through single-cell RNA-sequencing of the irradiated mouse intestine, we find that p53 target genes are specifically enriched in stem cells of the regenerating epithelium, including revival stem cells that promote animal survival after GI damage. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce revival stem cells. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells that is controlled by an Mdm2-mediated negative feedback loop. These results suggest that p53 suppresses severe radiation-indued GI injury by promoting intestinal epithelial cell reprogramming. One-Sentence Summary: After severe radiation injury to the intestine, transient p53 activity induces revival stem cells to promote regeneration.

6.
bioRxiv ; 2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37066384

RESUMO

The adaptive immune system plays an essential anti-tumor role through immunosurveillance and response to immunotherapies. Characterizing phenotypic features and mechanisms of dysfunction of tumor-specific T cell populations may uncover novel immunotherapeutic targets and biomarkers of response. To study tumor-specific T cell responses in vivo, a tumor model must express a known neoantigen. While transplant models with known neoantigen expression are widely available, autochthonous tumor models in which the tumor coevolves with the immune system are limited. In this study, we combined CRISPR/Cas9 and sleeping beauty transposase technology to develop an autochthonous orthotopic murine sarcoma model with oncogenic KrasG12D, functionally impaired p53, and expression of known MHCI and MHCII sarcoma neoantigens. Using MHC tetramer flow cytometry, we identified a tumor-specific immune response in the peripheral blood as early as 10 days after tumor induction leading to tumor clearance. Tumors developed at high penetrance after co-depletion of CD8 and CD4 T cells, but depletion of either CD8 or CD4 T cells alone was insufficient to permit tumor growth. These results suggest that CD8 and CD4 T cells can independently contribute to immunosurveillance leading to clearance of sarcomas expressing MHCI and MHCII neoantigens.

7.
Radiat Res ; 197(3): 0, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34724704

RESUMO

Delayed radiation myelopathy is a rare, but significant late side effect from radiation therapy that can lead to paralysis. The cellular and molecular mechanisms leading to delayed radiation myelopathy are not completely understood but may be a consequence of damage to oligodendrocyte progenitor cells and vascular endothelial cells. Here, we aimed to determine the contribution of endothelial cell damage to the development of radiation-induced spinal cord injury using a genetically defined mouse model in which endothelial cells are sensitized to radiation due to loss of the tumor suppressor p53. Tie2Cre; p53FL/+ and Tie2Cre; p53FL/- mice, which lack one and both alleles of p53 in endothelial cells, respectively, were treated with focal irradiation that specifically targeted the lumbosacral region of the spinal cord. The development of hindlimb paralysis was followed for up to 18 weeks after either a 26.7 Gy or 28.4 Gy dose of radiation. During 18 weeks of follow-up, 83% and 100% of Tie2Cre; p53FL/- mice developed hindlimb paralysis after 26.7 and 28.4 Gy, respectively. In contrast, during this period only 8% of Tie2Cre; p53FL/+ mice exhibited paralysis after 28.4 Gy. In addition, 8 weeks after 28.4 Gy the irradiated spinal cord from Tie2Cre; p53FL/- mice showed a significantly higher fractional area positive for the neurological injury marker glial fibrillary acidic protein (GFAP) compared with the irradiated spinal cord from Tie2Cre; p53FL/+ mice. Together, our findings show that deletion of p53 in endothelial cells sensitizes mice to the development of delayed radiation myelopathy indicating that endothelial cells are a critical cellular target of radiation that regulates myelopathy.


Assuntos
Traumatismos da Medula Espinal/radioterapia , Animais , Relação Dose-Resposta à Radiação , Células Endoteliais , Feminino , Proteína Glial Fibrilar Ácida/efeitos da radiação , Humanos , Masculino , Camundongos , Lesões Experimentais por Radiação , Radiação Ionizante , Medula Espinal/efeitos dos fármacos , Fatores de Tempo , Proteína Supressora de Tumor p53/efeitos da radiação
8.
Radiat Res ; 196(6): 686-689, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34644390

RESUMO

Exposure to high dose radiation causes life-threatening acute and delayed effects. Defining the mechanisms of lethal radiation-induced acute toxicity of gastrointestinal and hematopoietic tissues are critical steps to identify drug targets to mitigate and protect against the acute radiation syndrome (ARS). For example, one rational approach would be to design pharmaceuticals that block cell death pathways to preserve tissue integrity in radiation-sensitive organ systems including the gastrointestinal tract and hematopoietic compartment. A previous study reported that the inflammasome pathway, which mediates inflammatory cell death through pyroptosis, promotes ARS. However, we show that mice lacking the inflammatory executioner caspases, caspase-1 and caspase-11, are not protected from ARS when compared directly to littermates expressing caspase-1 and caspase-11. These results suggest that alternative pathways will need to be targeted by drugs that successfully mitigate and protect against the ARS.


Assuntos
Síndrome Aguda da Radiação/enzimologia , Caspase 1/metabolismo , Caspases Iniciadoras/metabolismo , Inflamassomos/metabolismo , Animais , Sistema Hematopoético/efeitos da radiação , Inflamassomos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Piroptose/efeitos da radiação
9.
Cancer Res ; 81(19): 4939-4948, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34385184

RESUMO

Chromosomal translocations generate oncogenic fusion proteins in approximately one-third of sarcomas, but how these proteins promote tumorigenesis is not well understood. Interestingly, some translocation-driven cancers exhibit dramatic clinical responses to therapy, such as radiotherapy, although the precise mechanism has not been elucidated. Here we reveal a molecular mechanism by which the fusion oncoprotein FUS-CHOP promotes tumor maintenance that also explains the remarkable sensitivity of myxoid liposarcomas to radiation therapy. FUS-CHOP interacted with chromatin remodeling complexes to regulate sarcoma cell proliferation. One of these chromatin remodelers, SNF2H, colocalized with FUS-CHOP genome-wide at active enhancers. Following ionizing radiation, DNA damage response kinases phosphorylated the prion-like domain of FUS-CHOP to impede these protein-protein interactions, which are required for transformation. Therefore, the DNA damage response after irradiation disrupted oncogenic targeting of chromatin remodelers required for FUS-CHOP-driven sarcomagenesis. This mechanism of disruption links phosphorylation of the prion-like domain of an oncogenic fusion protein to DNA damage after ionizing radiation and reveals that a dependence on oncogenic chromatin remodeling underlies sensitivity to radiation therapy in myxoid liposarcoma. SIGNIFICANCE: Prion-like domains, which are frequently translocated in cancers as oncogenic fusion proteins that drive global epigenetic changes, confer sensitivity to radiation via disruption of oncogenic interactions.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Radiação Ionizante , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos da radiação , Montagem e Desmontagem da Cromatina , Sequenciamento de Cromatina por Imunoprecipitação , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Fusão Oncogênica/química , Fosforilação/efeitos da radiação , Ligação Proteica , Proteína FUS de Ligação a RNA/química , Sarcoma/etiologia , Sarcoma/metabolismo , Sarcoma/patologia , Fator de Transcrição CHOP/química , Translocação Genética
10.
Cancer Res ; 81(14): 3777-3790, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34035082

RESUMO

Mouse models of radiation-induced thymic lymphoma are widely used to study the development of radiation-induced blood cancers and to gain insights into the biology of human T-cell lymphoblastic leukemia/lymphoma. Here we aimed to identify key oncogenic drivers for the development of radiation-induced thymic lymphoma by performing whole-exome sequencing using tumors and paired normal tissues from mice with and without irradiation. Thymic lymphomas from irradiated wild-type (WT), p53+/-, and KrasLA1 mice were not observed to harbor significantly higher numbers of nonsynonymous somatic mutations compared with thymic lymphomas from unirradiated p53-/- mice. However, distinct patterns of recurrent mutations arose in genes that control the Notch1 signaling pathway based on the mutational status of p53. Preferential activation of Notch1 signaling in p53 WT lymphomas was also observed at the RNA and protein level. Reporter mice for activation of Notch1 signaling revealed that total-body irradiation (TBI) enriched Notch1hi CD44+ thymocytes that could propagate in vivo after thymocyte transplantation. Mechanistically, genetic inhibition of Notch1 signaling in immature thymocytes prevented formation of radiation-induced thymic lymphoma in p53 WT mice. Taken together, these results demonstrate a critical role of activated Notch1 signaling in driving multistep carcinogenesis of thymic lymphoma following TBI in p53 WT mice. SIGNIFICANCE: These findings reveal the mutational landscape and key drivers in murine radiation-induced thymic lymphoma, a classic animal model that has been used to study radiation carcinogenesis for over 70 years.


Assuntos
Sequenciamento do Exoma/métodos , Linfoma/induzido quimicamente , Receptor Notch1/metabolismo , Neoplasias do Timo/induzido quimicamente , Proteína Supressora de Tumor p53/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos
11.
Radiother Oncol ; 157: 155-162, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33545252

RESUMO

BACKGROUND AND PURPOSE: Late cardiac toxicity is a major side effect of radiation therapy (RT) for breast cancer. We developed and characterized a mouse model of radiation-induced heart disease that mimics the dose, fractionation, and beam arrangement of left breast and chest wall RT. MATERIAL AND METHODS: Female wild-type (C57BL6/J) and atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice (on a C57BL/6J background) on regular chow were treated with 2 Gy × 25 fractions of partial-heart irradiation via opposed tangential beams to the left chest wall. The changes in myocardial perfusion and cardiac function of C57BL/6J mice were examined by single-photon emission computed tomography (SPECT) and echocardiography, respectively. In addition to SPECT and echocardiography, the formation of calcified plaques and changes in cardiac function of ApoE-/- mice were examined by dual-energy microCT (DE-CT) and pressure-volume (PV) loop analysis, respectively. The development of myocardial fibrosis was examined by histopathology. RESULTS: Compared to unirradiated controls, irradiated C57BL/6J mice showed no significant changes by SPECT or echocardiography up to 18 months after 2 Gy × 25 partial-heart irradiation even though irradiated mice exhibited a modest increase in myocardial fibrosis. For ApoE-/- mice, 2 Gy × 25 partial-heart irradiation did not cause significant changes by SPECT, DE-CT, or echocardiography. However, PV loop analysis revealed a significant decrease in load-dependent systolic and diastolic function measures including cardiac output, dV/dtmax and dV/dt min 12 months after RT. CONCLUSIONS: Following clinically relevant doses of partial-heart irradiation in C57BL/6J and ApoE-/- mice, assessment with noninvasive imaging modalities such as echocardiography, SPECT, and DE-CT yielded no evidence of decreased myocardial perfusion and cardiac dysfunction related to RT. However, invasive hemodynamic assessment with PV loop analysis indicated subtle, but significant, changes in cardiac function of irradiated ApoE-/- mice. PV loop analysis may be useful for future preclinical studies of radiation-induced heart disease, especially if subtle changes in cardiac function are expected.


Assuntos
Coração , Tomografia Computadorizada de Emissão de Fóton Único , Animais , Fracionamento da Dose de Radiação , Ecocardiografia , Feminino , Coração/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos C57BL
12.
Science ; 368(6487): 181-186, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32273467

RESUMO

Embryonic development is a complex process that is unamenable to direct observation. In this study, we implanted a window to the mouse uterus to visualize the developing embryo from embryonic day 9.5 to birth. This removable intravital window allowed manipulation and high-resolution imaging. In live mouse embryos, we observed transient neurotransmission and early vascularization of neural crest cell (NCC)-derived perivascular cells in the brain, autophagy in the retina, viral gene delivery, and chemical diffusion through the placenta. We combined the imaging window with in utero electroporation to label and track cell division and movement within embryos and observed that clusters of mouse NCC-derived cells expanded in interspecies chimeras, whereas adjacent human donor NCC-derived cells shrank. This technique can be combined with various tissue manipulation and microscopy methods to study the processes of development at unprecedented spatiotemporal resolution.


Assuntos
Embrião de Mamíferos/citologia , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Microscopia Intravital/métodos , Crista Neural , Animais , Encéfalo/embriologia , Encéfalo/fisiologia , Divisão Celular , Movimento Celular , Quimera/embriologia , Quimera/fisiologia , Eletroporação , Feminino , Técnicas de Transferência de Genes , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica , Crista Neural/irrigação sanguínea , Crista Neural/citologia , Crista Neural/embriologia , Placenta/fisiologia , Gravidez , Retina/embriologia , Retina/fisiologia , Transmissão Sináptica , Útero
13.
Health Phys ; 119(3): 315-321, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32175929

RESUMO

The Radiation and Nuclear Countermeasures Program at the National Institute of Allergy and Infectious Diseases (NIAID) mandated that medical countermeasures for treating Acute Radiation Syndrome (ARS) must have efficacy when administered at least 24 h after radiation exposure. At this time point, many cells within key target tissues, such as the hematopoietic system and the gastrointestinal (GI) tract, will already be dead. Therefore, drugs that promote the regeneration of surviving cells may improve outcomes. The serine/threonine kinase glycogen synthase kinase-3 (GSK-3) regulates stem and progenitor cell self-renewal and regeneration in the hematopoietic and GI compartments. We tested inhibition of GSK-3ß by SB216763 24 h after total body irradiation (TBI) and sub-total body irradiation (SBI). Here, we show that subcutaneous administration of SB216763 promotes the regeneration of surviving hematopoietic stem/progenitor cells (HSPCs), including myeloid progenitor cells, and improves survival of C57Bl/6 male mice when administered 24 h after TBI. However, these results were not recapitulated in female C57Bl/6 animals, suggesting a sex difference in GSK-3ß signaling in HSPCs. Subcutaneous administration of SB216763 in male mice stimulated activation of Sox2 transcription but failed to induce Sox2 transcription in female C57Bl/6 mice. Using TCF/lef-GFP reporter mice, we examined Wnt signaling in HSPCs of irradiated male and female mice treated with SB216763. GSK-3 inhibition elevated Wnt reporter activity in HSPCs isolated from male but not female mice. SB216763 did not mitigate hematopoietic ARS in males or females of a second strain of wild-type mice, C3H. In addition, administration of SB216763 did not mitigate hematopoietic ARS beyond the currently available standard approved therapy of ciprofloxacin and granulocyte-colony stimulating factor (G-CSF) in male C57Bl/6 mice. Further, SB216763 did not mitigate GI-ARS after SBI in C57Bl/6 male mice. The lack of efficacy in both sexes and multiple strains of mice indicate that SB216763 is not suitable for further drug development as a mitigator of ARS. Our studies demonstrate that activation of Wnt signaling in HSPCs promotes hematopoietic regeneration following radiation exposure, and targeting this pathway downstream of GSK-3ß may mitigate ARS in a sex- and strain-independent manner.


Assuntos
Síndrome Aguda da Radiação/prevenção & controle , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Hematopoese/efeitos da radiação , Indóis/uso terapêutico , Maleimidas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Protetores contra Radiação/uso terapêutico , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/enzimologia , Medula Óssea/efeitos da radiação , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Hematopoese/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores Sexuais , Especificidade da Espécie
14.
Appetite ; 146: 104509, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31698015

RESUMO

BACKGROUND: Maternal feeding styles have been associated with children's eating behaviors and obesity risk. Few works have identified maternal feeding styles using a multi-method person-centered approach. OBJECTIVES: (1) To identify maternal feeding styles using a person-centered multi-method approach, and (2) to examine the association of child weight status with maternal feeding styles. METHODS: Participants were low-income mother-child dyads (N = 255) (mean child age 5.9 years) from the United States. Mothers completed questionnaires and participated in a semi-structured interview. Interview transcripts were reliably coded for constructs of child feeding including beliefs, goals, and concerns. Family mealtime video recordings were reliably coded for feeding behaviors. Child anthropometrics were measured. Latent class analysis (LCA) was used to determine empirically-driven typologies of maternal feeding styles. Chi-square analyses tested the association of maternal feeding styles with child overweight or obese (vs. not) weight status. RESULTS: Two maternal feeding styles were identified by LCA which we term "High Coercive Control" (27% child overweight/obese) and "Low Coercive Control" (55% child overweight/obese). High Coercive Control mothers were more likely to believe their child was too thin, self-reported being more demanding in feeding and pressuring the child to eat, worried more about their child not eating enough and were observed to use more bribery. Low Coercive Control mothers were concerned about their child eating too much, and were less likely to self-report engaging in pressuring or restricting feeding behaviors. CONCLUSIONS: The findings suggest that although there is a "feeding style" characterized by substantial control, this style was most common among mothers of thinner children. The mothers of children with overweight/obesity were primarily characterized by engaging in the "recommended" feeding behaviors and being appropriately concerned about their child's risk for excess weight.


Assuntos
Comportamento Infantil/psicologia , Comportamento Alimentar/psicologia , Refeições/psicologia , Poder Familiar/psicologia , Pobreza/psicologia , Adulto , Antropometria , Peso Corporal , Pré-Escolar , Coerção , Feminino , Humanos , Análise de Classes Latentes , Masculino , Relações Mãe-Filho , Mães , Obesidade Pediátrica/psicologia , Projetos de Pesquisa , Fatores de Risco , Inquéritos e Questionários , Estados Unidos
15.
Nat Commun ; 10(1): 5647, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31827103

RESUMO

Intravital microscopy is a powerful technique to observe dynamic processes with single-cell resolution in live animals. No intravital window has been developed for imaging the colon due to its anatomic location and motility, although the colon is a key organ where the majority of microbiota reside and common diseases such as inflammatory bowel disease, functional gastrointestinal disorders, and colon cancer occur. Here we describe an intravital murine colonic window with a stabilizing ferromagnetic scaffold for chronic imaging, minimizing motion artifacts while maximizing long-term survival by preventing colonic obstruction. Using this setup, we image fluorescently-labeled stem cells, bacteria, and immune cells in live animal colons. Furthermore, we image nerve activity via calcium imaging in real time to demonstrate that electrical sacral nerve stimulation can activate colonic enteric neurons. The simple implantable apparatus enables visualization of live processes in the colon, which will open the window to a broad range of studies.


Assuntos
Colo/diagnóstico por imagem , Microscopia Intravital/métodos , Imagem Óptica/métodos , Animais , Movimento Celular , Colo/microbiologia , Corantes Fluorescentes/química , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/química , Células-Tronco/citologia
16.
Radiat Res ; 192(3): 258-266, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31265788

RESUMO

Exposure of the gastrointestinal (GI) tract to ionizing radiation can cause acute and delayed injury. However, critical cellular targets that regulate the development of radiation-induced GI injury remain incompletely understood. Here, we investigated the role of vascular endothelial cells in controlling acute and delayed GI injury after total-abdominal irradiation (TAI). To address this, we used genetically engineered mice in which endothelial cells are sensitized to radiation due to the deletion of the tumor suppressor p53. Remarkably, we found that VE-cadherin-Cre; p53FL/FL mice, in which both alleles of p53 are deleted in endothelial cells, were not sensitized to the acute GI radiation syndrome, but these mice were highly susceptible to delayed radiation enteropathy. Histological examination indicated that VE-cadherin-Cre; p53FL/FL mice that developed delayed radiation enteropathy had severe vascular injury in the small intestine, which was manifested by hemorrhage, loss of microvessels and tissue hypoxia. In addition, using dual-energy CT imaging, we showed that VE-cadherin-Cre; p53FL/FL mice had a significant increase in vascular permeability of the small intestine in vivo 28 days after TAI. Together, these findings demonstrate that while sensitization of endothelial cells to radiation does not exacerbate the acute GI radiation syndrome, it is sufficient to promote the development of late radiation enteropathy.


Assuntos
Células Endoteliais/patologia , Células Endoteliais/efeitos da radiação , Intestinos/patologia , Intestinos/efeitos da radiação , Tolerância a Radiação , Animais , Permeabilidade Capilar/efeitos da radiação , Hipóxia Celular/efeitos da radiação , Células Endoteliais/metabolismo , Deleção de Genes , Camundongos , Fatores de Tempo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
17.
JCI Insight ; 4(13)2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31112524

RESUMO

Cancer development is influenced by hereditary mutations, somatic mutations due to random errors in DNA replication, or external factors. It remains unclear how distinct cell-intrinsic and -extrinsic factors impact oncogenesis within the same tissue type. We investigated murine soft tissue sarcomas generated by oncogenic alterations (KrasG12D activation and p53 deletion), carcinogens (3-methylcholanthrene [MCA] or ionizing radiation), and in a novel model combining both factors (MCA plus p53 deletion). Whole-exome sequencing demonstrated distinct mutational signatures in individual sarcoma cohorts. MCA-induced sarcomas exhibited high mutational burden and predominantly G-to-T transversions, while radiation-induced sarcomas exhibited low mutational burden and a distinct genetic signature characterized by C-to-T transitions. The indel to substitution ratio and amount of gene copy number variations were high for radiation-induced sarcomas. MCA-induced tumors generated on a p53-deficient background showed the highest genomic instability. MCA-induced sarcomas harbored mutations in putative cancer-driver genes that regulate MAPK signaling (Kras and Nf1) and the Hippo pathway (Fat1 and Fat4). In contrast, radiation-induced sarcomas and KrasG12Dp53-/- sarcomas did not harbor recurrent oncogenic mutations, rather they exhibited amplifications of specific oncogenes: Kras and Myc in KrasG12Dp53-/- sarcomas, and Met and Yap1 for radiation-induced sarcomas. These results reveal that different initiating events drive oncogenesis through distinct mechanisms.


Assuntos
Carcinogênese/genética , Neoplasias Experimentais/genética , Neoplasias Induzidas por Radiação/genética , Oncogenes/genética , Sarcoma/genética , Animais , Carcinogênese/efeitos da radiação , Carcinógenos/toxicidade , Análise Mutacional de DNA , Instabilidade Genômica/efeitos da radiação , Humanos , Metilcolantreno/toxicidade , Camundongos , Neoplasias Experimentais/induzido quimicamente , Oncogenes/efeitos da radiação , Proteínas Proto-Oncogênicas p21(ras)/genética , Sarcoma/induzido quimicamente , Proteína Supressora de Tumor p53/genética , Sequenciamento do Exoma
18.
Int J Radiat Oncol Biol Phys ; 102(5): 1569-1576, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30056081

RESUMO

PURPOSE: The delivery of radiation therapy to cure gastrointestinal (GI) cancers is often limited by normal tissue toxicity of the GI tract. Studies using genetically engineered mice have demonstrated an essential role of the cyclin-dependent kinase inhibitor p21 in protecting against GI acute radiation syndrome (GI-ARS). Here, we examined the impact of the Food and Drug Administration-approved, selective, cyclin-dependent kinase 4/6 inhibitor palbociclib (PD-0332991) on the development of GI-ARS induced by single-dose versus fractionated radiation in mice. METHODS AND MATERIALS: For the single-dose radiation study, C57BL/6J mice were treated with palbociclib or vehicle 28 and 4 hours before subtotal body irradiation (SBI). For the fractionated radiation study, C57BL/6J mice were exposed to fractionated SBI for 5 consecutive days. These mice were treated with palbociclib or vehicle either 28 and 4 hours before the first dose of irradiation or 4 hours before the first, third, and fifth doses of irradiation. RESULTS: Our data indicate that treatment with palbociclib before, but not after, a single fraction of SBI significantly ameliorated GI-ARS, improved the integrity of the GI barrier, and increased the number of surviving crypts in the small intestine. In addition, palbociclib did not protect tumor cell lines from radiation in vitro. In contrast to the results from the single-dose exposure, treatment with palbociclib before 5 daily fractions of SBI did not prevent GI-ARS. Moreover, we unexpectedly observed that GI-ARS was exacerbated in mice treated with palbociclib before and during 5 daily fractions of SBI. CONCLUSIONS: Our results demonstrate that treatment with palbociclib before a single dose of SBI protects mice from GI-ARS. In contrast, treatment with palbociclib before and during 5 daily fractions of SBI exacerbates GI-ARS in mice. These results emphasize the importance of conducting preclinical studies of radioprotectors with single-dose and fractionated radiation therapy.


Assuntos
Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Fracionamento da Dose de Radiação , Trato Gastrointestinal/efeitos dos fármacos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Lesões Experimentais por Radiação/tratamento farmacológico , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Trato Gastrointestinal/efeitos da radiação , Intestinos/efeitos dos fármacos , Intestinos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Lesões Experimentais por Radiação/etiologia , Lesões Experimentais por Radiação/patologia
19.
Radiat Res ; 189(6): 627-633, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29634408

RESUMO

Exposure to high doses of ionizing radiation can cause lethal injury to normal tissue, thus inducing acute radiation syndrome. Acute radiation syndrome is caused by depletion of bone marrow cells (hematopoietic syndrome) and irreparable damage to the epithelial cells in the gastrointestinal tract (gastrointestinal syndrome). Although radiation initiates apoptosis in the hematopoietic and gastrointestinal compartments within the first few hours after exposure, alternative mechanisms of cell death may contribute to injury in these radiosensitive tissues. In this study, we utilized mice lacking a critical regulator of necroptosis, receptor interacting protein 3 (RIP3) kinase, to characterize the role of RIP3 in normal tissue toxicity after irradiation. Our results suggest that RIP3-mediated signaling is not a critical driver of acute radiation syndrome.


Assuntos
Síndrome Aguda da Radiação/enzimologia , Síndrome Aguda da Radiação/genética , Técnicas de Inativação de Genes , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Síndrome Aguda da Radiação/patologia , Animais , Apoptose/efeitos da radiação , Hematopoese/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Necrose/etiologia , Transdução de Sinais/efeitos da radiação
20.
J Mol Endocrinol ; 56(3): R99-R114, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26831511

RESUMO

Steroid hormone receptors (SRs) are heavily posttranslationally modified by the reversible addition of a variety of molecular moieties, including phosphorylation, acetylation, methylation, SUMOylation, and ubiquitination. These rapid and dynamic modifications may be combinatorial and interact (i.e. may be sequential, complement, or oppose each other), creating a vast array of uniquely modified receptor subspecies that allow for diverse receptor behaviors that enable highly sensitive and context-dependent hormone action. For example, in response to hormone or growth factor membrane-initiated signaling events, posttranslational modifications (PTMs) to SRs alter protein-protein interactions that govern the complex process of promoter or gene-set selection coupled to transcriptional repression or activation. Unique phosphorylation events allow SRs to associate or disassociate with specific cofactors that may include pioneer factors and other tethering partners, which specify the resulting transcriptome and ultimately change cell fate. The impact of PTMs on SR action is particularly profound in the context of breast tumorigenesis, in which frequent alterations in growth factor-initiated signaling pathways occur early and act as drivers of breast cancer progression toward endocrine resistance. In this article, with primary focus on breast cancer relevance, we review the mechanisms by which PTMs, including reversible phosphorylation events, regulate the closely related SRs, glucocorticoid receptor and progesterone receptor, allowing for precise biological responses to ever-changing hormonal stimuli.


Assuntos
Neoplasias da Mama/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de Glucocorticoides/metabolismo , Receptores de Progesterona/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Prognóstico , Isoformas de Proteínas , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Receptores de Progesterona/química , Receptores de Progesterona/genética , Transdução de Sinais , Estresse Fisiológico , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...