Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cancers (Basel) ; 16(3)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38339228

RESUMEN

Despite recent advances in cancer therapy, ovarian cancer remains the most lethal gynecological cancer worldwide, making it crucial and of the utmost importance to establish novel therapeutic strategies. Adjuvant radiotherapy has been assessed historically, but its use was limited by intestinal toxicity. We recently established the role of Limosilactobacillus reuteri in releasing IL-22 (LR-IL-22) as an effective radiation mitigator, and we have now assessed its effect in an ovarian cancer mouse model. We hypothesized that an LR-IL-22 gavage would enable intestinal radioprotection by modifying the tumor microenvironment and, subsequently, improving overall survival in female C57BL/6MUC-1 mice with widespread abdominal syngeneic 2F8cis ovarian cancer. Herein, we report that the LR-IL-22 gavage not only improved overall survival in mice when combined with a PD-L1 inhibitor by inducing differential gene expression in irradiated stem cells but also induced PD-L1 protein expression in ovarian cancer cells and mobilized CD8+ T cells in whole abdomen irradiated mice. The addition of LR-IL-22 to a combined treatment modality with fractionated whole abdomen radiation (WAI) and systemic chemotherapy and immunotherapy regimens can facilitate a safe and effective protocol to reduce tumor burden, increase survival, and improve the quality of life of a locally advanced ovarian cancer patient.

2.
In Vivo ; 37(6): 2421-2432, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37905617

RESUMEN

BACKGROUND/AIM: Patients with radiation sensitive Fanconi anemia (FA) are presenting with cancers of the oral cavity, oropharynx, and other anatomic locations. MATERIALS AND METHODS: Animal models for cancer in FA mice used orthotopic tumors from wild type mice. We derived a cancer cell line from Fanca-/- mice by topical application of the chemical carcinogen dimethyl benzanthracene (DMBA). RESULTS: A Fanca-/- mouse rhabdomyosarcoma was derived from a Fanca-/- (129/Sv) mouse. The in vitro clonogenic survival of the Fanca-/- clone 6 cancer cell line was consistent with the FA genotype. Transplanted tumors demonstrated hypoxic centers surrounded by senescent cells. CONCLUSION: This Fanca-/- mouse syngeneic cancer should provide a valuable resource for discovery and development of new normal tissue radioprotectors for patients with FA and cancer.


Asunto(s)
Anemia de Fanconi , Neoplasias , Humanos , Ratones , Animales , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Línea Celular , Carcinógenos/toxicidad , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética
3.
Cell Death Discov ; 9(1): 252, 2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460469

RESUMEN

Cellular senescence is involved in the development of pulmonary fibrosis as well as in lung tissue repair and regeneration. Therefore, a strategy of removal of senescent cells by senolytic drugs may not produce the desired therapeutic result. Previously we reported that tyrosine kinase Fgr is upregulated in ionizing irradiation-induced senescent cells. Inhibition of Fgr reduces the production of profibrotic proteins by radiation-induced senescent cells in vitro; however, a mechanistic relationship between senescent cells and radiation-induced pulmonary fibrosis (RIPF) has not been established. We now report that senescent cells from the lungs of mice with RIPF, release profibrotic proteins for target cells and secrete chemotactic proteins for marrow cells. The Fgr inhibitor TL02-59, reduces this release of profibrotic chemokines from the lungs of RIPF mice, without reducing numbers of senescent cells. In vitro studies demonstrated that TL02-59 abrogates the upregulation of profibrotic genes in target cells in transwell cultures. Also, protein arrays using lung fibroblasts demonstrated that TL02-59 inhibits the production of chemokines involved in the migration of macrophages to the lung. In thoracic-irradiated mice, TL02-59 prevents RIPF, significantly reduces levels of expression of fibrotic gene products, and significantly reduces the recruitment of CD11b+ macrophages to the lungs. Bronchoalveolar lavage (BAL) cells from RIPF mice show increased Fgr and other senescent cell markers including p16. In human idiopathic pulmonary fibrosis (IPF) and in RIPF, Fgr, and other senescent cell biomarkers are increased. In both mouse and human RIPF, there is an accumulation of Fgr-positive proinflammatory CD11b+ macrophages in the lungs. Thus, elevated levels of Fgr in lung senescent cells upregulate profibrotic gene products, and chemokines that might be responsible for macrophage infiltration into lungs. The detection of Fgr in senescent cells that are obtained from BAL during the development of RIPF may help predict the onset and facilitate the delivery of medical countermeasures.

4.
Cancers (Basel) ; 15(6)2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36980556

RESUMEN

Irradiation can be an effective treatment for ovarian cancer, but its use is limited by intestinal toxicity. Thus, strategies to mitigate toxicity are important and can revitalize the current standard of care. We previously established that LR-IL-22 protects the intestine from WAI. We now hypothesize that LR-IFN-ß is an effective radiation protector and mitigator and is rapidly cleared from the digestive tract, making it an option for intestinal radioprotection. We report that the gavage of LR-IFN-ß during WAI provides improved intestinal barrier integrity and significantly preserves the numbers of Lgr5+GFP+ intestinal stem cells, improving survival. The rapid clearance of the genetically engineered probiotic from the digestive tract renders it a safe and feasible radiation mitigator. Therefore, the above genetically engineered probiotic is both a feasible and effective radiation mitigator that could potentially revolutionize the management of OC patients. Furthermore, the subsequent addition of platinum/taxane-based chemotherapy to the combination of WAI and LR-IFN-ß should reduce tumor volume while protecting the intestine and should improve the overall survival in OC patients.

5.
In Vivo ; 37(2): 506-518, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36881075

RESUMEN

BACKGROUND/AIM: The earliest cellular and molecular biologic changes in the esophagus that lead to esophageal cancer were evaluated in a mouse model. We correlated numbers of senescent cells with the levels of expression of potentially carcinogenic genes in sorted side population (SP) cells containing esophageal stem cells and non-stem cells in the non-side population cells in the 4-nitroquinolone oxide (NQO)-treated esophagus. MATERIALS AND METHODS: We compared stem cells with non-stem cells from the esophagus of mice treated with the chemical carcinogen 4-NQO (100 µg/ml) in drinking water. We also compared gene expression in human esophagus samples treated with 4-NQO (100 µg/ml media) to non-treated samples. We separated and quantitated the relative levels of expression of RNA using RNAseq analysis. We identified senescent cells by luciferase imaging of p16+/LUC mice and senescent cells in excised esophagus from tdTOMp16+ mice. RESULTS: A significant increase in the levels of RNA for oncostatin-M was found in senescent cells of the esophagus from 4-NQO-treated mice and human esophagus in vitro. CONCLUSION: Induction of OSM in chemically-induced esophageal cancer in mice correlates with the appearance of senescent cells.


Asunto(s)
Neoplasias Esofágicas , Nitroquinolinas , Humanos , Animales , Ratones , Carcinógenos , Óxidos , Mutágenos , Neoplasias Esofágicas/inducido químicamente , Neoplasias Esofágicas/genética , ARN , Oncostatina M
6.
Int J Mol Sci ; 23(10)2022 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-35628427

RESUMEN

(1) Background: The systemic administration of therapeutic agents to the intestine including cytokines, such as Interleukin-22 (IL-22), is compromised by damage to the microvasculature 24 hrs after total body irradiation (TBI). At that time, there is significant death of intestinal microvascular endothelial cells and destruction of the lamina propria, which limits drug delivery through the circulation, thus reducing the capacity of therapeutics to stabilize the numbers of Lgr5+ intestinal crypt stem cells and their progeny, and improve survival. By its direct action on intestinal stem cells and their villus regeneration capacity, IL-22 is both an ionizing irradiation protector and mitigator. (2) Methods: To improve delivery of IL-22 to the irradiated intestine, we gavaged Lactobacillus-reuteri as a platform for the second-generation probiotic Lactobacillus-reuteri-Interleukin-22 (LR-IL-22). (3) Results: There was effective radiation mitigation by gavage of LR-IL-22 at 24 h after intestinal irradiation. Multiple biomarkers of radiation damage to the intestine, immune system and bone marrow were improved by LR-IL-22 compared to the gavage of control LR or intraperitoneal injection of IL-22 protein. (4) Conclusions: Oral administration of LR-IL-22 is an effective protector and mitigator of intestinal irradiation damage.


Asunto(s)
Limosilactobacillus reuteri , Probióticos , Protección Radiológica , Células Endoteliales , Interleucinas , Mucosa Intestinal/metabolismo , Intestinos , Interleucina-22
7.
Radiat Res ; 198(1): 89-105, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35446961

RESUMEN

Oral administration (gavage) of a second-generation probiotic, Lactobacillus reuteri (L. reuteri), that releases interleukin-22 (LR-IL-22) at 24 h after total-body irradiation (TBI) mitigates damage to the intestine. We determined that LR-IL-22 also mitigates partial-body irradiation (PBI) and whole-abdomen irradiation (WAI). Irradiation can be an effective treatment for ovarian cancer, but its use is limited by intestinal toxicity. Strategies to mitigate toxicity are important and can revitalize this modality to treat ovarian cancer. In the present studies, we evaluated whether LR-IL-22 facilitates fractionated WAI in female C57BL/6 mice with disseminated ovarian cancer given a single fraction of either 15.75 Gy or 19.75 Gy or 4 daily fractions of 6 Gy or 6.5 Gy. Mice receiving single or multiple administrations of LR-IL-22 during WAI showed improved intestinal barrier integrity (P = 0.0167), reduced levels of radiation-induced intestinal cytokines including KC/CXCL1 (P = 0.002) and IFN-γ (P = 0.0024), and reduced levels of plasma, Eotaxin/CCL11 (P = 0.0088). LR-IL-22 significantly preserved the numbers of Lgr5+GFP+ intestinal stem cells (P = 0.0010) and improved survival (P < 0.0343). Female C57BL/6MUC-1 mice with widespread abdominal syngeneic 2F8cis ovarian cancer that received LR-IL-22 during 6.5 Gy WAI in 4 fractions had reduced tumor burden, less intestinal toxicity, and improved 30-day survival. Furthermore, LR-IL-22 facilitated WAI when added to Paclitaxel and Carboplatin chemotherapy and further increased survival. Oral administration (gavage) of LR-IL-22 is a potentially valuable intestinal radioprotector, which can facilitate therapeutic WAI for widespread intra-abdominal ovarian cancer.


Asunto(s)
Limosilactobacillus reuteri , Neoplasias Ováricas , Abdomen , Animales , Carcinoma Epitelial de Ovario , Femenino , Humanos , Interleucinas , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias Ováricas/patología , Neoplasias Ováricas/radioterapia , Interleucina-22
8.
In Vivo ; 35(6): 3053-3066, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34697137

RESUMEN

BACKGROUND/AIM: The role of senescence and bone marrow-derived cells in silica-induced pulmonary fibrosis is unknown. MATERIALS AND METHODS: C57BL/6HNsd, p16+/LUC, and tdTOMp16+ mice were intratracheally injected with 200 mg/kg crystalline silica or irradiated (20 Gy) to the thoracic cavity and followed for the development of lung fibrosis. RESULTS: The p16+/LUC mice demonstrated senescence by day 7 after silica exposure. C57BL/6 mice exposed to silica demonstrated upregulation of p16, p21, and tyrosine kinase Fgr by day 7, whereas thoracic irradiation induced p21 and Fgr by day 50 and p16 by day 110. Silica exposed GFP+ bone marrow chimeric C57BL/6 mice demonstrated senescent cells and gfp+/Fgr+ monocyte/macrophages in the lungs on day 21. The Fgr inhibitor TL02-59 abrogated monocyte/macrophages recruitment in in vitro transwell experiments. CONCLUSION: Both silica and radiation exposure induce senescence and upregulate tyrosine kinase Fgr for the recruitment of bone marrow-derived monocyte/macrophages and the development of pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar , Dióxido de Silicio , Animales , Médula Ósea , Senescencia Celular , Pulmón , Macrófagos , Ratones , Ratones Endogámicos C57BL , Monocitos , Fibrosis Pulmonar/inducido químicamente , Dióxido de Silicio/toxicidad
9.
Radiat Res ; 196(3): 235-249, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34087939

RESUMEN

We defined the time course of ionizing radiation-induced senescence in lung compared to bone marrow of p16+/LUC mice in which the senescence-induced biomarker (p16) is linked to a luciferase reporter gene. Periodic in situ imaging revealed increased luciferase activity in the lungs of 20 Gy thoracic irradiated, but not 8 Gy total-body irradiated (TBI) mice beginning at day 75 and increasing to day 170. In serial sections of explanted lungs, senescent cells appeared in the same areas as did fibrosis in the 20 Gy thoracic irradiated, but not the 8 Gy TBI group. Lungs from 8 Gy TBI mice at one year did show increased RNA levels for p16, p21, p19 and TGF-ß. Individual senescent cells in 20 Gy irradiated mouse lung included those with epithelial, endothelial, fibroblast and hematopoietic cell biomarkers. Rare senescent cells in the lungs of 8 Gy TBI mice at one year were of endothelial phenotype. Long-term bone marrow cultures (LTBMCs) were established at either day 60 or one year after 8 Gy TBI. In freshly removed marrow at both times after irradiation, there were increased senescent cells. In LTBMCs, there were increased senescent cells in both weekly harvested single cells and in colonies of multilineage hematopoietic progenitor cells producing CFU-GEMM (colony forming unit-granulocyte, erythrocyte, monocyte/macrophage, mega-karyocyte) that were formed in secondary cultures when these single cells were plated in semisolid media. LTBMCs from TBI mice produced fewer CFU-GEMM; however, the relative percentage of senescent cell-containing colonies was increased as measured by both p16-luciferase and ß-galactosidase. Therefore, 20 Gy thoracic radiation, as well as 8 Gy TBI, induces senescent cells in the lungs. With bone marrow, 8 Gy TBI induced senescence in both hematopoietic cells and in colony-forming progenitors. The p16+/LUC mouse strain provides a valuable system in which to compare the kinetics of radiation-induced senescence between organs in vivo, and to evaluate the potential role of senescent cells in irradiation pulmonary fibrosis.


Asunto(s)
Médula Ósea/efectos de la radiación , Senescencia Celular/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Células Madre Hematopoyéticas/efectos de la radiación , Pulmón/efectos de la radiación , Células Madre Multipotentes/efectos de la radiación , Irradiación Corporal Total/efectos adversos , Animales , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Genes p16 , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/etiología , Traumatismos Experimentales por Radiación/etiología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transgenes
10.
Radiat Res ; 193(5): 435-450, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32134361

RESUMEN

Mitigation of total-body irradiation (TBI) in C57BL/6 mice by two drugs, which target apoptosis and necroptosis respectively, increases survival compared to one drug alone. Here we investigated whether the biomarker (signature)directed addition of a third anti-ferroptosis drug further mitigated TBI effects. C57BL/6NTac female mice (30-33 g) received 9.25 Gy TBI, and 24 h or later received JP4-039 (20 mg/kg), necrostatin-1 (1.65 mg/kg) and/or lipoxygenase-15 inhibitor (baicalein) (50 mg/kg) in single-, dual- or three-drug regimens. Some animals were sacrificed at days 0, 1, 2, 3, 4 or 7 postirradiation, while the majority in each group were maintained beyond 30 days. For those mice sacrificed at the early time points, femur bone marrow, intestine (ileum), lung and blood plasma were collected and analyzed for radiation-induced and mitigator-modified levels of 33 pro-inflammatory and stress response proteins. Each single mitigator administered [JP4-039 (24 h), necrostatin-1 (48 h) or baicalein (24 h)] improved survival at day 30 after TBI to 25% (P = 0.0432, 0.2816 or 0.1120, respectively) compared to 5% survival of 9.25 Gy TBI controls. Mice were administered the drug individually based on weight (mg/kg). Drug vehicles comprised 30% cyclodextrin for JP4-039 and baicalein, and 10% Cremphor-EL/10% ethanol/80% water for necrostatin-1; thus, dual-vehicle controls were also tested. The dual-drug combinations further enhanced survival: necrostatin-1 (delayed to 72 h) with baicalein 40% (P = 0.0359); JP4-039 with necrostatin-1 50% (P = 0.0062); and JP4-039 with baicalein 60% (P = 0.0064). The three-drug regimen, timed to signature directed evidence of onset after TBI of each death pathway in marrow and intestine, further increased the 30-day survival to 75% (P = 0.0002), and there was optimal normalization to preirradiation levels of inflammatory cytokine and stress response protein levels in plasma, intestine and marrow. In contrast, lung protein levels were minimally altered by 9.25 Gy TBI or mitigators over 7 days. Significantly, elevated intestinal proteins at day 7 after TBI were reduced by necrostatin-1-containing regimens; however, normalization of plasma protein levels at day 7 required the addition of JP4-039 and baicalein. These findings indicate that mitigator targeting to three distinct cell death pathways increases survival after TBI.


Asunto(s)
Apoptosis/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Necroptosis/efectos de los fármacos , Protectores contra Radiación/farmacología , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis/efectos de la radiación , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Médula Ósea/efectos de la radiación , Citocinas/metabolismo , Interacciones Farmacológicas , Femenino , Ferroptosis/efectos de la radiación , Íleon/efectos de los fármacos , Íleon/metabolismo , Íleon/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Necroptosis/efectos de la radiación , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/prevención & control , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Tiempo
11.
In Vivo ; 34(1): 33-38, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31882460

RESUMEN

BACKGROUND/AIM: Radiation mitigator, GS-nitroxide, JP4-039, was evaluated for mitigation of total body irradiation (TBI) in Fanconi anemia (FA) Fancd2-/- (129/Sv), Fancg-/- (B6), and Fanca-/- (129/Sv) mice. MATERIALS AND METHODS: JP4-039 dissolved in 30% 2-hydroxypropyl-ß-cyclodextrin was injected intramuscularly 24 h after total body irradiation (9.25 Gy) into Fanca-/-, Fancd2-/- and Fancg-/- mice. Irradiation survival curves were performed in vitro using bone marrow stromal cell lines derived from Fanca-/-, Fancd2-/- and Fancg-/- mice. RESULTS: FA mice demonstrate genotype specific differences in TBI mitigation by JP4-039. Radiation effects in derived bone marrow stromal cell lines in vitro were mitigated by drugs that block apoptosis, but not necroptosis or ferroptosis. CONCLUSION: FA mouse models are valuable for elucidating DNA repair pathways in cell and tissue responses to TBI, and the role of drugs that target distinct cell death pathways.


Asunto(s)
Anemia de Fanconi/genética , Óxidos de Nitrógeno/farmacología , Protectores contra Radiación/farmacología , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/efectos de la radiación , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Línea Celular , Modelos Animales de Enfermedad , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Genotipo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/genética , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética
12.
In Vivo ; 33(3): 675-688, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31028184

RESUMEN

Background/Aim: The cause of fatal neuromuscular amyotrophic lateral sclerosis (ALS) is not known. Materials and Methods: Ninety-day-old superoxide-dismutase-1 G93A (SOD1 G93A ) mice demonstrating level 1 paralysis, received 9.0 Gy total body irradiation (TBI) from a cesium source at 340 cGy per minute, and intravenous transplantation with 1×10 6 C57BL/6 green fluorescent protein (GFP)+ donor bone marrow cells. Results: Paralysis-free survival was prolonged in TBI and bone marrow-transplanted SOD1 G93A mice from 100 to over 250 days (p=0.0018). Other mice transplanted with SOD1 G93A marrow or marrow treated with the free-radical scavenger MMS350 showed no therapeutic effect. GFP+ macrophage-2 (M2) microglial cells of bone marrow origin, were seen at sites of degenerating anterior horn motor neurons. SOD1 G93A mice had a disruption in the blood-brain barrier permeability which was reversed by marrow transplant from C57BL/6 mice. SOD1 G93A marrow showed unexpected robust hematopoiesis in vitro, and radioresistance. Conclusion: After TBI, M2 microglial cells from transplanted donor marrow extended the paralysis-free interval in SOD1 G93A mice.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Microglía/metabolismo , Mutación , Superóxido Dismutasa-1/genética , Sustitución de Aminoácidos , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/terapia , Animales , Barrera Hematoencefálica/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Expresión Génica , Supervivencia de Injerto , Hematopoyesis/genética , Ratones , Ratones Transgénicos , Microglía/inmunología , Tolerancia a Radiación/genética , Superóxido Dismutasa-1/metabolismo , Quimera por Trasplante
13.
In Vivo ; 33(2): 303-311, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30804107

RESUMEN

AIM: To demonstrate that Fanconi anemia complementation group D2-deficient (Fancd2-/-) hematopoietic progenitor cell lines can be transformed by transfection with a plasmid containing either the E6 or E7 oncogene of human papillomavirus (HPV) to generate malignant plasmacytoma-inducing cell lines. MATERIALS AND METHODS: In order to determine whether a single HPV type 16 (HPV16) oncogene induced malignant transformation, Fancd2-/- and Fancd2+/+ interleukin 3 (IL3)-dependent hematopoietic progenitor cell lines were transfected with plasmids containing E6 or E7 oncogene, or control empty plasmid. RESULTS: Fancd2-/- but not Fancd2+/+ cells were transformed into malignant IL3-independent cells by both E6, and E7 oncogenes, but not by empty plasmid. Hematopoietic cell lines and tumors induced by Fancd2-/- E6 and Fancd2-/- E7 cell lines were positive for each respective HPV RNA and protein. CONCLUSION: A single HPV16 oncogene is adequate to produce malignant transformation of Fancd2-/- hematopoietic cells.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Células Madre Hematopoyéticas/virología , Interleucina-3/genética , Línea Celular Tumoral , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidad , Humanos , Proteínas Oncogénicas Virales/genética , Oncogenes/genética , Proteínas E7 de Papillomavirus/genética , Plásmidos/genética , Proteínas Represoras/genética , Transfección/métodos
14.
Radiat Res ; 191(2): 139-153, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30499383

RESUMEN

We quantitated age-related accumulation of senescent cells in irradiated Fanconi anemia (FA) (Fanca-/- mouse cell lines in vitro, and monitored the effect of continuous administration (via drinking water) of the water-soluble radiation mitigator, MMS350, on tissues in vivo over one year after 7.5 Gy total-body irradiation (TBI). Irradiated Fanca-/- mouse bone marrow stromal cell lines showed increased numbers of beta-galactosidase- and p21-positive senescent cells compared to Fanca+/+ cell lines, which was reduced by MMS350. One week after 7.5 Gy TBI, Fanca-/- mice showed increased numbers of senescent cells in spleen compared to Fanca+/+ controls, decreased bone marrow cellularity, failure of explanted bone marrow to proliferate in vitro to form a hematopoietic microenvironment and no detectable single stromal cell cloning capacity. There was no detectable amelioration by MMS350 administration at one week. In contrast, one year post-TBI, Fanca-/- mice demonstrated fewer senescent cells in brain and spleen compared to Fanca+/+ controls. While Fanca-/- mouse bone marrow stromal cells explanted one year post-TBI still failed to proliferate in vitro, continuous oral administration of 400 µ M, MMS350 in drinking water restored explanted stromal cell proliferation. The data indicate that continuous administration of MMS350 modulated several properties of TBI-accelerated aging in Fanca-/- mice as well as control mice, and support further study of MMS350 as a modulator of radiation late effects.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Éteres Cíclicos/administración & dosificación , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de la radiación , Protectores contra Radiación/administración & dosificación , Sulfóxidos/administración & dosificación , Irradiación Corporal Total , Administración Oral , Animales , Éteres Cíclicos/farmacología , Femenino , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Protectores contra Radiación/farmacología , Sulfóxidos/farmacología , Microambiente Tumoral
15.
Radiat Res ; 189(6): 560-578, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29584588

RESUMEN

Squamous cell carcinomas of the head and neck are appearing with increased frequency in both marrow transplanted and non-transplanted Fanconi anemia (FA) patients. FA patients commonly display radiosensitivity of epithelial tissues, complicating effective radiotherapy. Fancd2-/- mice (C57BL/6J and 129/Sv background) demonstrate epithelial tissue sensitivity to single-fraction or fractionated irradiation to the head and neck and distant marrow suppression (abscopal effect), both ameliorated by intraoral administration of the mitochondrial-targeted antioxidant, GS-nitroxide, JP4-039. We now report that mice of two other FA genotypes, Fancg-/- (B6) and the most prevalent human genotype Fanca-/- (129/Sv), also demonstrate: 1. reduced longevity of hematopoiesis in long-term bone marrow cultures; 2. radiosensitivity of bone marrow stromal cell lines; and 3. head and neck radiation-induced severe mucositis and abscopal suppression of distant marrow hematopoiesis. Intraoral administration of JP4-039/F15, but not non-mitochondrial-targeted 4-amino-Tempo/F15 or F15 alone, prior to each radiation treatment ameliorated both local and abscopal radiation effects. Head and neck irradiated TGF-ß-resistant SMAD3-/- (129/Sv) mice and double-knockout SMAD3-/- Fancd2-/- (129/Sv) mice treated daily with TGF-ß receptor antagonist, LY364947, still displayed abscopal bone marrow suppression, implicating a non-TGF-ß mechanism. Thus, amelioration of both local normal tissue radiosensitivity and distant marrow suppression by intraoral administration of JP4-039 in Fancg-/- and Fanca-/- mice supports a clinical trial of this locally administered normal tissue radioprotector and mitigator during head and neck irradiation in FA patients.


Asunto(s)
Médula Ósea/efectos de los fármacos , Neoplasias de Cabeza y Cuello/radioterapia , Mucositis/tratamiento farmacológico , Óxidos de Nitrógeno/administración & dosificación , Óxidos de Nitrógeno/farmacología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Protectores contra Radiación/farmacología , Administración Oral , Animales , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Proteína del Grupo de Complementación A de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación G de la Anemia de Fanconi/deficiencia , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Interleucina-3/metabolismo , Ratones , Mitomicina/farmacología , Mucositis/metabolismo , Mucositis/patología , Óxidos de Nitrógeno/uso terapéutico , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/efectos de la radiación , Protectores contra Radiación/administración & dosificación , Protectores contra Radiación/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Factor de Crecimiento Transformador beta/metabolismo
16.
In Vivo ; 31(3): 313-320, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28438857

RESUMEN

BACKGROUND/AIM: The polycythemia form of Friend leukemia virus (FVP) causes splenomegaly and lethal erythroleukemia in Fv-2s-susceptible mouse strains. We sought to determine whether the hematopoietic stem cell (HSC) pool was expanded in Fv-2r-resistant mice. MATERIALS AND METHODS: The 120-day bone marrow transplantation competitive repopulation assay was used to determine whether FVP-infected Fv-2r C57BL/6 mice demonstrated expansion of the HSC pool compared to the pool of committed hematopoietic progenitor cells in the same marrow assayed in vitro. RESULTS: There was a significant expansion of committed hematopoietic progenitors observed in virus-infected Fv-2s FVB mice, but not Fv-2r C57BL/6 mice. Furthermore, Fv-2r mice showed no detectable expansion of either committed hematopoietic progenitor cells or the multipotential stem cell pool by competitive repopulation assay. CONCLUSION: Friend virus disease in Fv-2s mice is associated with expansion of committed hematopoietic progenitors. Fv-2r mice show no expansion of either committed progenitor or pluripotential stem cell numbers.


Asunto(s)
Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Infecciones por Retroviridae/patología , Infecciones Tumorales por Virus/patología , Animales , Médula Ósea/virología , Femenino , Virus de la Leucemia Murina de Friend/patogenicidad , Células Madre Hematopoyéticas/virología , Leucemia Eritroblástica Aguda/patología , Leucemia Eritroblástica Aguda/virología , Leucemia Experimental/patología , Leucemia Experimental/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Retroviridae/virología , Bazo/patología , Bazo/virología , Esplenomegalia/patología , Esplenomegalia/virología , Infecciones Tumorales por Virus/virología
17.
In Vivo ; 31(2): 159-168, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28358695

RESUMEN

BACKGROUND/AIM: Total-body irradiation and/or administration of chemotherapy drugs in bone marrow transplantation induce cytokines that can suppress engraftment. Fanconi Anemia (FA) patients have a hyperactive responsiveness to the inhibitory cytokine, transforming growth factor-beta (TGF-ß). Small molecule radiation mitigator drugs, JP4-039 and MMS350, were evaluated for suppression of irradiation or drug-induced TGF-ß. MATERIALS AND METHODS: In vivo induction of TGF-ß by total-body ionizing irradiation (TBI), L-phenylalanine mustard (L-PAM), busulfan or fludarabine, was quantified. In parallel, mitigator drug amelioration of TGF-ß induction in FA D2-/- (FANCD2-/-) mouse bone marrow, was studied in vitro. Tissue culture medium, cell lysates, and mouse plasma were analyzed for TGF-ß levels. RESULTS: Induction of TGF-ß levels in FANCD2-/- and FANCD2+/+ mice and in mouse bone marrow were modulated by both JP4-039 and MMS350. CONCLUSION: Bone marrow transplantation in FA recipients may benefit from administration of small molecule agents that suppress TGF-ß induction.


Asunto(s)
Médula Ósea/efectos de los fármacos , Éteres Cíclicos/farmacología , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/radioterapia , Óxidos de Nitrógeno/farmacología , Sulfóxidos/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Western Blotting , Médula Ósea/metabolismo , Busulfano/farmacología , Línea Celular , Células Cultivadas , Quimioterapia/métodos , Anemia de Fanconi/metabolismo , Melfalán/farmacología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Agonistas Mieloablativos/farmacología , Protectores contra Radiación/farmacología , Técnicas de Cultivo de Tejidos , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/genética , Vidarabina/análogos & derivados , Vidarabina/farmacología , Irradiación Corporal Total/métodos
18.
Radiat Res ; 185(2): 134-50, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26789701

RESUMEN

We evaluated normal tissue specific radioprotection of the oral cavity in radiosensitive Fanconi Anemia (FA) Fancd2(-/-) mice with orally established tumors using mitochondrial-targeted GS-nitroxide (JP4-039). Adult (10-12 weeks old) Fancd2(+/+), Fancd2(+/-) and Fancd2(-/-) mice (C57BL/6 background) and subgroups with orally established TC-1 epithelial cell tumors received a single fraction of 28 Gy or four daily fractions of 8 Gy to the head and neck. Subgroups received JP4-039 in F15 emulsion (F15/JP4-039; 0.4 mg/mouse), 4-amino-Tempo in F15 emulsion (F15/4-amino-Tempo; 0.2 mg/mouse) or F15 emulsion alone prior to each irradiation. Oral mucosa of Fancd2(-/-) mice showed baseline elevated RNA transcripts for Sod2, p53, p21 and Rad51 (all P < 0.0012) and suppressed levels of Nfkb and Tgfb, (all P < 0.0020) compared with Fancd2(+/+) mice. The oral mucosa in tumor-bearing mice of all genotypes showed decreased levels of p53 and elevated Tgfb and Gadd45a (P ≤ 0.0001 for all three genotypes). Intraoral F15/JP4-039, but not F15/4-amino-Tempo, modulated radiation-induced normal tissue transcript elevation, ameliorated mucosal ulceration and reduced the depletion of antioxidant stores in oral cavity tissue of all genotypes, but did not radioprotect tumors. Mitochondrial targeting makes F15/JP4-039 an effective normal tissue radioprotector for Fancd2(-/-) mice, as well as wild-type mice.


Asunto(s)
Mitocondrias/efectos de los fármacos , Mitocondrias/efectos de la radiación , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/efectos de la radiación , Neoplasias de la Boca/radioterapia , Óxidos de Nitrógeno/administración & dosificación , Administración Oral , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucosa Bucal/patología , Neoplasias de la Boca/patología , Órganos en Riesgo , Tolerancia a Radiación/efectos de los fármacos , Protectores contra Radiación/administración & dosificación , Resultado del Tratamiento
19.
In Vivo ; 28(5): 699-708, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25189880

RESUMEN

AIM: To determine if the small-molecule radioprotector GS-nitroxide, JP4-039, improved hematopoiesis in long-term bone marrow cultures (LTBMCs), explanted marrow from in vivo drug-treated C57BL/6NTac mice was maintained in JP4-039 for 25 weeks. Hematopoietic cell production and radiobiology of derived stromal cell lines was measured. MATERIALS AND METHODS: Groups of LTBMCs were established from mouse groups. Stromal cell lines were established from the adherent layer of JP4-039-treated and untreated control groups. RESULTS: LTBMCs maintained in JP4-039 exhibited increased production of total non-adherent and 7-day and 14-day hematopoietic colony-forming cells. Stromal cell lines derived from JP4-039-treated cultures were radioresistant in vitro, demonstrated a distinct squamous/epithelial morphology and overexpressed Nrf2, Ctgf, Lox, Tlr1, collagen 1a, Brd3, and Brd4. CONCLUSION: Chronic treatment of bone marrow cultures and derived stromal cell lines with JP4-039 was non-toxic, and conferred resistance to oxidative stress.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de la radiación , Óxidos de Nitrógeno/farmacología , Tolerancia a Radiación/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de la radiación , Línea Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Perfilación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Tolerancia a Radiación/genética , Transcripción Genética
20.
In Vivo ; 28(4): 441-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24982208

RESUMEN

AIM: We measured long-term hematopoiesis in continuous bone marrow cultures derived from Toll-like receptor-4 (Tlr4(-/-))(C57BL/6J) mice. MATERIALS AND METHODS: We measured hematopoiesis in vitro over 27 weeks in long-term bone marrow cultures from Tlr4(-/-) and control mice, and irradiation-induced pulmonary fibrosis in mice irradiated to 20 Gy to the thorax. RESULTS: There was a significant increase in the duration of hematopoiesis in long-term bone marrow cultures from Tlr4(-/-) mice in production of total non-adherent cells and day 7 and day 14 multi-lineage colony-forming cells. The histology of bone marrow hematopoietic and stromal cell lines was indistinguishable between different mouse strains. There was no detectable late irradiation pulmonary fibrosis in Tlr4(-/-) mice. CONCLUSION: Homozygous deletion of both alleles of Tlr4, encoding for an inflammatory mediator receptor, improves the duration of hematopoiesis in vitro and reduces irradiation-induced lung fibrosis.


Asunto(s)
Eliminación de Gen , Hematopoyesis/genética , Hematopoyesis/efectos de la radiación , Fibrosis Pulmonar/etiología , Radiación , Receptor Toll-Like 4/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Ensayo de Unidades Formadoras de Colonias , Modelos Animales de Enfermedad , Homocigoto , Ratones , Ratones Noqueados , Fibrosis Pulmonar/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA