Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 722
Filtrer
1.
In Vivo ; 38(4): 1719-1730, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936939

RÉSUMÉ

BACKGROUND/AIM: ClFdA is a second-generation antineoplastic agent that has demonstrated significant anticancer activity, particularly against acute lymphoblastic leukemia and has been shown to have radiosensitizing activity. The aim of the study was to explore the genotoxic, cytotoxic and radiosensitizing effects of clofarabine (ClFdA) on bone marrow cells (BMCs), normoblasts and leukocytes of mice in vivo. MATERIALS AND METHODS: Cytotoxicity was determined by the reduction in reticulocytes (RET), and genotoxicity was determined by the induction of micronucleated reticulocytes (MN-RET) in the peripheral blood and by DNA break induction in leukocytes determined by single-cell gel electrophoresis (SCGE). The radiosensitizing capacity of ClFdA was determined in leukocytes and BMCs by SCGE. RESULTS: Two mechanisms of MN-RET induction were identified according to the antecedents, that could be due to inhibition of DNA synthesis and demethylation of G-C regions, and subsequent chromosome fragility. ClFdA cytotoxicity causes two contiguous peaks, an early peak that seems to inhibit MN-RET induction and a second peak that seems to be caused by ribonucleotide reductase (RR) and/or DNA synthesis inhibitions. ClFdA induced early DNA damage in noncycling leukocytes, and also radiosensitizes leukocytes immediately after treatment. ClFdA-ionizing radiation (IR) causes two time-dependent episodes of DNA damage, the latest after 80 min triggers a major breakage of DNA. In terms of the number of damaged cells, leukocytes and BMCs are similarly sensitive to ionizing radiation; BMCs are slightly more sensitive than leukocytes to ClFdA, but BMCs are doubly sensitive to combined treatment. CONCLUSION: ClFdA causes early DNA damage and radiosensitivity in non-proliferating leukocytes, which rules out the most favored hypotheses of the participation of RR and DNA polymerase inhibition.


Sujet(s)
Clofarabine , Altération de l'ADN , Leucocytes , Radiosensibilisants , Animaux , Clofarabine/pharmacologie , Souris , Radiosensibilisants/pharmacologie , Leucocytes/effets des médicaments et des substances chimiques , Leucocytes/effets des radiations , Altération de l'ADN/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des radiations , Arabinonucléosides/pharmacologie , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/effets des radiations , Cellules de la moelle osseuse/métabolisme , Nucléotides adényliques/pharmacologie , Mâle , Réticulocytes/effets des médicaments et des substances chimiques , Réticulocytes/effets des radiations , Antinéoplasiques/pharmacologie , Tests de micronucleus
2.
Nature ; 618(7966): 834-841, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37286599

RÉSUMÉ

Tumours most often arise from progression of precursor clones within a single anatomical niche. In the bone marrow, clonal progenitors can undergo malignant transformation to acute leukaemia, or differentiate into immune cells that contribute to disease pathology in peripheral tissues1-4. Outside the marrow, these clones are potentially exposed to a variety of tissue-specific mutational processes, although the consequences of this are unclear. Here we investigate the development of blastic plasmacytoid dendritic cell neoplasm (BPDCN)-an unusual form of acute leukaemia that often presents with malignant cells isolated to the skin5. Using tumour phylogenomics and single-cell transcriptomics with genotyping, we find that BPDCN arises from clonal (premalignant) haematopoietic precursors in the bone marrow. We observe that BPDCN skin tumours first develop at sun-exposed anatomical sites and are distinguished by clonally expanded mutations induced by ultraviolet (UV) radiation. A reconstruction of tumour phylogenies reveals that UV damage can precede the acquisition of alterations associated with malignant transformation, implicating sun exposure of plasmacytoid dendritic cells or committed precursors during BPDCN pathogenesis. Functionally, we find that loss-of-function mutations in Tet2, the most common premalignant alteration in BPDCN, confer resistance to UV-induced cell death in plasmacytoid, but not conventional, dendritic cells, suggesting a context-dependent tumour-suppressive role for TET2. These findings demonstrate how tissue-specific environmental exposures at distant anatomical sites can shape the evolution of premalignant clones to disseminated cancer.


Sujet(s)
Transformation cellulaire néoplasique , Cellules dendritiques , Leucémie aigüe myéloïde , Tumeurs cutanées , Peau , Rayons ultraviolets , Humains , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Cellules de la moelle osseuse/effets des radiations , Mort cellulaire/effets des radiations , Lignage cellulaire/génétique , Lignage cellulaire/effets des radiations , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Transformation cellulaire néoplasique/effets des radiations , Clones cellulaires/métabolisme , Clones cellulaires/anatomopathologie , Clones cellulaires/effets des radiations , Cellules dendritiques/métabolisme , Cellules dendritiques/anatomopathologie , Cellules dendritiques/effets des radiations , Leucémie aigüe myéloïde/étiologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Mutation/effets des radiations , Spécificité d'organe , Analyse de l'expression du gène de la cellule unique , Tumeurs cutanées/étiologie , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Rayons ultraviolets/effets indésirables , Peau/anatomopathologie , Peau/effets des radiations
3.
Wei Sheng Yan Jiu ; 51(1): 91-98, 2022 Jan.
Article de Chinois | MEDLINE | ID: mdl-35341500

RÉSUMÉ

OBJECTIVE: To investigate the radioprotective effect of gallic acid(GA) on mouse bone marrow cells. METHODS: Healthy male ICR mice were randomly divided into saline control group, GA control group, X-ray irradiation group and GA protection group, with 10 mice in each group. X-ray irradiation group and normal saline control group were given 0.01 mL/g normal saline gavage, GA control group and GA protection group were given 200 mg/kg GA(20 mg/mL) gavage once a day for 14 consecutive days. On the 15 th day, 4 X-ray irradiation groups and 4 GA protection groups were given one-time X-ray irradiation to the whole body of the mice, and the absorbed doses were 1.0, 2.0, 3.0 and 4.0 Gy, respectively. The saline control group and the GA control group were not irradiated. After irradiation, detected the whole blood catalase(CAT), superoxide dismutase(SOD), malondialdehyde(MDA) and micronucleus frequency of polychromatic erythrocyte in bone marrow(MN-PCE), and use flow cytometry to detect bone marrow cell cycle, early apoptosis rate and late apoptosis rate. RESULTS: The CAT activities in the serum of mice in the 1.0, 2.0, 3.0 and 4.0 Gy GA protection groups were 2.13, 1.74, 1.49 and 1.15 U/mL, respectively, which were significantly increased compared with the corresponding X-ray irradiation group(P<0.01). SOD activities were 184.69, 156.92, 139.17 and 107.15 U/mL, which were significantly increased compared with the corresponding X-ray irradiation group(P<0.01). The contents of MDA were 3.92, 4.20, 6.32 and 9.31 nmol/mL, which were significantly lower than those of the corresponding X-ray irradiation group(P<0.05, P<0.01). The bone marrow MN-PCE rate of the mice in the 1.0, 2.0, 3.0 and 4.0 Gy GA protection groups were 4.35‰, 8.00‰, 12.90‰ and 3.80‰, respectively, which were significantly lower than those in the corresponding X-ray irradiation group(P<0.01). The proportions of G_0/G_1 phase cells of bone marrow cells in the 1.0, 2.0, 3.0 and 4.0 Gy GA protection group were 81.00%, 86.28%, 92.04% and 93.15%, respectively, which were significantly reduced compared with the corresponding X-ray irradiation group(P<0.01). The proportions of G_2/M phase cells were 4.51%, 3.05%, 2.35% and 1.81%, which were significantly increased compared with the corresponding X-ray irradiation group(P<0.05, P<0.01). The proportions of S phase cells were 15.32%, 11.36%, 5.96% and 4.92%, which were significantly increased compared with the corresponding X-ray irradiation group(P<0.01). The early apoptosis rate of bone marrow cells of mice in the 1.0, 2.0, 3.0 and 4.0 Gy GA protection groups were 3.32%, 8.96%, 12.11% and 2.26%, respectively, which were significantly reduced compared with the corresponding X-ray irradiation group(P<0.01). The late apoptosis rates of bone marrow cells were 7.21%, 11.73%, 17.11% and 19.36%, which were significantly reduced compared with the corresponding X-ray irradiation group(P<0.05, P<0.01). CONCLUSION: GA can reduce the oxidative damage, DNA damage and bone marrow cell cycle arrest of the bone marrow cells of radiation-damaged mice, inhibit the apoptosis of bone marrow cells, and have radioprotective effects on the bone marrow cells of mice.


Sujet(s)
Cellules de la moelle osseuse , Acide gallique , Animaux , Moelle osseuse/effets des radiations , Cellules de la moelle osseuse/effets des radiations , Altération de l'ADN , Acide gallique/pharmacologie , Mâle , Souris , Souris de lignée ICR
4.
Cancer Lett ; 524: 172-181, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34688844

RÉSUMÉ

The influence of high-linear energy transfer (LET) particle radiation on the functionalities of mesenchymal stromal cells (MSCs) is largely unknown. Here, we analyzed the effects of proton (1H), helium (4He), carbon (12C) and oxygen (16O) ions on human bone marrow-MSCs. Cell cycle distribution and apoptosis induction were examined by flow cytometry, and DNA damage was quantified using γH2AX immunofluorescence and Western blots. Relative biological effectiveness values of MSCs amounted to 1.0-1.1 for 1H, 1.7-2.3 for 4He, 2.9-3.4 for 12C and 2.6-3.3 for 16O. Particle radiation did not alter the MSCs' characteristic surface marker pattern, and MSCs maintained their multi-lineage differentiation capabilities. Apoptosis rates ranged low for all radiation modalities. At 24 h after irradiation, particle radiation-induced ATM and CHK2 phosphorylation as well as γH2AX foci numbers returned to baseline levels. The resistance of human MSCs to high-LET irradiation suggests that MSCs remain functional after exposure to moderate doses of particle radiation as seen in normal tissues after particle radiotherapy or during manned space flights. In the future, in vivo models focusing on long-term consequences of particle irradiation on the bone marrow niche and MSCs are needed.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie/génétique , Checkpoint kinase 2/génétique , Histone/génétique , Cellules souches mésenchymateuses/effets des radiations , Cellules souches/effets des radiations , Médecine aérospatiale , Apoptose/génétique , Apoptose/effets des radiations , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Cellules de la moelle osseuse/effets des radiations , Carbone/effets indésirables , Cycle cellulaire/génétique , Cycle cellulaire/effets des radiations , Lignage cellulaire/génétique , Lignage cellulaire/effets des radiations , Cytométrie en flux , Régulation de l'expression des gènes/effets des radiations , Hélium/effets indésirables , Humains , Cellules souches mésenchymateuses/métabolisme , Oxygène/effets indésirables , Protons/effets indésirables , Vol spatial , Cellules souches/métabolisme
5.
Biomed Pharmacother ; 146: 112350, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-34952740

RÉSUMÉ

This study explored the radioprotective effects and possible underlying mechanisms of KR-31831 against radiation-induced injury in a mouse model. KR-31831 (30 and 50 mg/kg) was administered to mice 24 h and 30 min before exposure to a single lethal or sublethal dose of whole-body irradiation (WBI) (7 or 4 Gy, respectively). These animals were then evaluated for changes in mortality, various hematological and biochemical parameters, and histological features in response to these treatments. In addition, RNA sequencing was used to profile the radiation-induced transcriptomic response in the bone marrow cells. The results showed that KR-31831 dose-dependently prolonged the 30-day survival period and prevented damage to radiation-sensitive organs, such as the intestine and testis, in response to WBI. Damage to the hematopoietic system was also notably improved in the KR-31831-treated mice, as evidenced by an increase in bone marrow and peripheral blood cells, as well as recovery of the histopathological characteristics of the bone marrow. These protective effects were achieved, at least in part, via the suppression of radiation-induced increases in apoptotic cell death and erythropoietin levels in the plasma. Furthermore, the gene expression profiles of the bone marrow cells of the WBI-treated mice suggested that KR-31831 upregulates the expression of the genes involved in regulating apoptosis and modulating the immune response, both of which are required for protecting the bone marrow. These results suggest the potential therapeutic efficacy of KR-31831 for protection against radiation-induced injury.


Sujet(s)
Benzopyranes/usage thérapeutique , Imidazoles/usage thérapeutique , Lésions radiques/traitement médicamenteux , Radioprotecteurs/usage thérapeutique , Irradiation corporelle totale/effets indésirables , Animaux , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/effets des radiations , Intestins/effets des médicaments et des substances chimiques , Intestins/effets des radiations , Mâle , Souris de lignée C57BL , Lésions radiques/génétique , Testicule/effets des médicaments et des substances chimiques , Testicule/effets des radiations , Transcriptome/effets des médicaments et des substances chimiques
6.
Int J Mol Sci ; 22(18)2021 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-34576128

RÉSUMÉ

Radiation-induced abscopal effect (RIAE) outside of radiation field is becoming more attractive. However, the underlying mechanisms are still obscure. This work investigated the deleterious effect of thoracic irradiation (Th-IR) on distant bone marrow and associated signaling factors by irradiating the right thorax of mice with fractionated doses (8 Gy × 3). It was found that this localized Th-IR increased apoptosis of bone marrow cells and micronucleus formation of bone marrow polychromatic erythrocytes after irradiation. Tandem mass tagging (TMT) analysis and ELISA assay showed that the concentrations of TNF-α and serum amyloid A (SAA) in the mice were significantly increased after Th-IR. An immunohistochemistry assay revealed a robust increase in SAA expression in the liver rather than in the lungs after Th-IR. In vitro experiments demonstrated that TNF-α induced SAA expression in mouse hepatoma Hepa1-6 cells, and these two signaling factors induced DNA damage in bone marrow mesenchymal stem cells (BMSCs) by increasing reactive oxygen species (ROS). On the other hand, injection with TNF-α inhibitor before Th-IR reduced the secretion of SAA and attenuated the abscopal damage in bone marrow. ROS scavenger NAC could also mitigated Th-IR/SAA-induced bone marrow damage in mice. Our findings indicated that Th-IR triggered TNF-α release from lung, which further promoted SAA secretion from liver in a manner of cascade reaction. Consequently, these signaling factors resulted in induction of abscopal damage on bone marrow of mice.


Sujet(s)
Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/effets des radiations , Fractionnement de la dose d'irradiation , Protéine amyloïde A sérique/métabolisme , Thorax/effets des radiations , Facteur de nécrose tumorale alpha/métabolisme , Acétylcystéine/pharmacologie , Animaux , Protéines du sang/métabolisme , Cycle cellulaire/effets des radiations , Altération de l'ADN , Piégeurs de radicaux libres/pharmacologie , Lésion pulmonaire/anatomopathologie , Mâle , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des radiations , Souris de lignée C57BL , Protéomique , Espèces réactives de l'oxygène/métabolisme
7.
Exp Mol Med ; 53(9): 1319-1331, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34497346

RÉSUMÉ

Although functional interplay between intestinal microbiota and distant sites beyond the gut has been identified, the influence of microbiota-derived metabolites on hematopoietic stem cells (HSCs) remains unclear. This study investigated the role of microbiota-derived lactate in hematopoiesis using mice deficient in G-protein-coupled receptor (Gpr) 81 (Gpr81-/-), an established lactate receptor. We detected significant depletion of total HSCs in the bone marrow (BM) of Gpr81-/- mice compared with heterogenic (Gpr81+/-) mice in a steady state. Notably, the expression levels of stem cell factor (SCF), which is required for the proliferation of HSCs, decreased significantly in leptin receptor-expressing (LepR+) mesenchymal stromal cells (MSCs) around the sinusoidal vessels of the BM from Gpr81-/- mice compared with Gpr81+/- mice. Hematopoietic recovery and activation of BM niche cells after irradiation or busulfan treatment also required Gpr81 signals. Oral administration of lactic acid-producing bacteria (LAB) activated SCF secretion from LepR+ BM MSCs and subsequently accelerated hematopoiesis and erythropoiesis. Most importantly, LAB feeding accelerated the self-renewal of HSCs in germ-free mice. These results suggest that microbiota-derived lactate stimulates SCF secretion by LepR+ BM MSCs and subsequently activates hematopoiesis and erythropoiesis in a Gpr81-dependent manner.


Sujet(s)
Hématopoïèse , Interactions hôte-microbes , Acide lactique/métabolisme , Microbiote , Récepteurs à la leptine/métabolisme , Facteur de croissance des cellules souches/métabolisme , Niche de cellules souches , Animaux , Marqueurs biologiques , Moelle osseuse/métabolisme , Moelle osseuse/effets des radiations , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/effets des radiations , Érythropoïèse , Cellules souches hématopoïétiques , Immunophénotypage , Souris , Souris knockout , Modèles biologiques , Probiotiques , Transduction du signal
8.
Environ Mol Mutagen ; 62(7): 422-427, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34296472

RÉSUMÉ

It is well-known that the cytotoxicity and mutagenic effects of high dose rate (HDR) ionizing radiation (IR) are increased by increasing the dose but less is known about the effects of chronic low dose rate (LDR). In vitro, we have shown that in addition to the immediate interaction of IR with DNA (the direct and indirect effects), low doses and chronic LDR exposure induce endogenous oxidative stress. During elevated oxidative stress, reactive oxygen species (ROS) react with DNA modifying its structure. Here, BL6 mice were exposed to IR at LDR and HDR and were then sacrificed 3 hours and 3 weeks after exposure to examine early and late effects of IR. The levels of micronuclei, MN, were determined in bone marrow cells. Our data indicate that the effects of 200 mGy on MN-induction are transient, but 500 and 1000 mGy (both HDR and LDR) lead to increased levels of MN up to 3 weeks after the exposure.


Sujet(s)
Cellules de la moelle osseuse/anatomopathologie , Rayons gamma/effets indésirables , Micronoyaux à chromosomes défectueux/effets des radiations , Irradiation corporelle totale/effets indésirables , Animaux , Cellules de la moelle osseuse/effets des radiations , Relation dose-effet des rayonnements , Femelle , Souris , Souris de lignée C57BL , Tests de micronucleus
9.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-34203224

RÉSUMÉ

Previous work pointed to a critical role of excessive production of reactive oxygen species (ROS) in increased radiation hematopoietic death in GFP mice. Meanwhile, enhanced antioxidant capability was not demonstrated in the mouse model of radio-induced adaptive response (RAR) using rescue of radiation hematopoietic death as the endpoint. ROS induction by ex vivo X-irradiation at a dose ranging from 0.1 to 7.5 Gy in the nucleated bone marrow cells was comparatively studied using GFP and wild type (WT) mice. ROS induction was also investigated in the cells collected from mice receiving a priming dose (0.5 Gy) efficient for RAR induction in WT mice. Significantly elevated background and increased induction of ROS in the cells from GFP mice were observed compared to those from WT mice. Markedly lower background and decreased induction of ROS were observed in the cells collected from WT mice but not GFP mice, both receiving the priming dose. GFP overexpression could alter background and induction of ROS by X-irradiation in hematopoietic cells. The results provide a reasonable explanation to the previous study on the fate of cells and mice after X-irradiation and confirm enhanced antioxidant capability in RAR. Investigations involving GFP overexpression should be carefully interpreted.


Sujet(s)
Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/effets des radiations , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/effets des radiations , Espèces réactives de l'oxygène/métabolisme , Rayons X/effets indésirables , Animaux , Relation dose-effet des rayonnements , Femelle , Souris , Souris de lignée C57BL
10.
Biomed Mater ; 16(5)2021 08 17.
Article de Anglais | MEDLINE | ID: mdl-34330118

RÉSUMÉ

In this study, we evaluated cardiomyogenic differentiation of electromechanically stimulated rat bone marrow-derived stem cells (rt-BMSCs) on an acellular bovine pericardium (aBP) and we looked at the functioning of this engineered patch in a rat myocardial infarct (MI) model. aBP was prepared using a detergent-based decellularization procedure followed by rt-BMSCs seeding, and electrical, mechanical, or electromechanical stimulations (3 millisecond pulses of 5 V cm-1at 1 Hz, 5% stretching) to enhance cardiomyogenic differentiation. Furthermore, the electromechanically stimulated patch was applied to the MI region over 3 weeks. After this period, the retrieved patch and infarct region were evaluated for the presence of calcification, inflammatory reaction (CD68), patch to host tissue cell migration, and structural sarcomere protein expressions. In conjunction with any sign of calcification, a higher number of BrdU-labelled cells, and a low level of CD68 positive cells were observed in the infarct region under electromechanically stimulated conditions compared with static conditions. More importantly, MHC, SAC, Troponin T, and N-cad positive cells were observed in both infarct region, and retrieved engineered patch after 3 weeks. In a clear alignment with other results, our developed acellular patch promoted the expression of cardiomyogenic differentiation factors under electromechanical stimulation. Our engineered patch showed a successful integration with the host tissue followed by the cell migration to the infarct region.


Sujet(s)
Matériaux biocompatibles , Stimulation électrique , Infarctus du myocarde , Myocarde , Ingénierie tissulaire/méthodes , Animaux , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/effets des radiations , Bovins , Différenciation cellulaire/effets des médicaments et des substances chimiques , Myocarde/cytologie , Myocarde/métabolisme , Myocytes cardiaques/cytologie , Péricarde/cytologie , Péricarde/transplantation , Rats , Cellules souches/cytologie , Cellules souches/effets des radiations
11.
Stem Cells Transl Med ; 10(7): 1095-1114, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33724714

RÉSUMÉ

Acute radiation syndrome (ARS) is the radiation toxicity that can affect the hematopoietic, gastrointestinal, and nervous systems upon accidental radiation exposure within a short time. Currently, there are no effective and safe approaches to treat mass population exposure to ARS. Our study aimed to evaluate the therapeutic potential of allogeneic adipose-derived stem cells (ASCs) for total body irradiation (TBI)-induced ARS and understand the underlying mitigation mechanism. We employed 9.25 Gy TBI dose to C57BL/6 mice and studied the effect of allogeneic ASCs on mice survival and regeneration of the hematopoietic system. Our results indicate that intraperitoneal-injected ASCs migrated to the bone marrow, rescued hematopoiesis, and improved the survival of irradiated mice. Our transwell coculture results confirmed the migration of ASCs to irradiated bone marrow and rescue hematopoietic activity. Furthermore, contact coculture of ASCs improved the survival and hematopoiesis of irradiated bone marrow in vitro. Irradiation results in DNA damage, upregulation of inflammatory signals, and apoptosis in bone marrow cells, while coculture with ASCs reduces apoptosis via activation of DNA repair and the antioxidation system. Upon exposure to irradiated bone marrow cells, ASCs secrete prosurvival and hematopoietic factors, such as GM-CSF, MIP1α, MIP1ß, LIX, KC, 1P-10, Rantes, IL-17, MCSF, TNFα, Eotaxin, and IP-10, which reduces oxidative stress and rescues damaged bone marrow cells from apoptosis. Our findings suggest that allogeneic ASCs therapy is effective in mitigating TBI-induced ARS in mice and may be beneficial for clinical adaptation to treat TBI-induced toxicities. Further studies will help to advocate the scale-up and adaptation of allogeneic ASCs as the radiation countermeasure.


Sujet(s)
Syndrome d'irradiation aigu , Apoptose , Cellules de la moelle osseuse/effets des radiations , Transplantation de cellules souches hématopoïétiques , Syndrome d'irradiation aigu/thérapie , Tissu adipeux/cytologie , Animaux , Hématopoïèse , Souris , Souris de lignée C57BL
12.
Mol Med Rep ; 23(6)2021 06.
Article de Anglais | MEDLINE | ID: mdl-33786622

RÉSUMÉ

The loosening and displacement of prostheses after dental implantation and arthroplasty is a substantial medical burden due to the complex correction surgery. Three­dimensional (3D)­printed porous titanium (pTi) alloy scaffolds are characterized by low stiffness, are beneficial to bone ingrowth, and may be used in orthopedic applications. However, for the bio­inert nature between host bone and implants, titanium alloy remains poorly compatible with osseointegration, especially in disease conditions, such as osteoporosis. In the present study, 3D­printed pTi scaffolds with ideal pore size and porosity matching the bone tissue, were combined with pulse electromagnetic fields (PEMF), an exogenous osteogenic induction stimulation, to evaluate osseointegration in osteoporosis. In vitro, external PEMF significantly improved osteoporosis­derived bone marrow mesenchymal stem cell proliferation and osteogenic differentiation on the surface of pTi scaffolds by enhancing the expression of alkaline phosphatase, runt­related transcription factor­2, osteocalcin, and bone morphogenetic protein­2. In vivo, Microcomputed tomography analysis and histological evaluation indicated the external PEMF markedly enhanced bone regeneration and osseointegration. This novel therapeutic strategy has potential to promote osseointegration of dental implants or artificial prostheses for patients with osteoporosis.


Sujet(s)
Alliages/composition chimique , Champs électromagnétiques , Ostéo-intégration , Ostéoporose/chirurgie , Impression tridimensionnelle , Ingénierie tissulaire/méthodes , Structures d'échafaudage tissulaires/composition chimique , Titane/composition chimique , Phosphatase alcaline/métabolisme , Animaux , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/physiologie , Cellules de la moelle osseuse/effets des radiations , Protéine morphogénétique osseuse de type 2/métabolisme , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Sous-unité alpha 1 du facteur CBF/métabolisme , Femelle , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/physiologie , Cellules souches mésenchymateuses/effets des radiations , Ostéocalcine/métabolisme , Lapins
13.
Sci Rep ; 11(1): 5653, 2021 03 11.
Article de Anglais | MEDLINE | ID: mdl-33707490

RÉSUMÉ

Exposure to moderate doses of ionizing radiation (IR), which is sufficient for causing skin injury, can occur during radiation therapy as well as in radiation accidents. Radiation-induced skin injury occasionally recovers, although its underlying mechanism remains unclear. Moderate-dose IR is frequently utilized for bone marrow transplantation in mice; therefore, this mouse model can help understand the mechanism. We had previously reported that bone marrow-derived cells (BMDCs) migrate to the epidermis-dermis junction in response to IR, although their role remains unknown. Here, we investigated the role of BMDCs in radiation-induced skin injury in BMT mice and observed that BMDCs contributed to skin recovery after IR-induced barrier dysfunction. One of the important mechanisms involved the action of CCL17 secreted by BMDCs on irradiated basal cells, leading to accelerated proliferation and recovery of apoptosis caused by IR. Our findings suggest that BMDCs are key players in IR-induced skin injury recovery.


Sujet(s)
Cellules de la moelle osseuse/anatomopathologie , Kératinocytes/anatomopathologie , Lésions radiques/anatomopathologie , Animaux , Cellules de la moelle osseuse/effets des radiations , Transplantation de moelle osseuse , Mouvement cellulaire/effets des radiations , Prolifération cellulaire/effets des radiations , Chimiokine CCL17/métabolisme , Derme/anatomopathologie , Derme/effets des radiations , Épiderme/anatomopathologie , Épiderme/effets des radiations , Délétion de gène , Cellules HaCaT , Humains , Kératinocytes/effets des radiations , Macrophages/effets des radiations , Souris de lignée C57BL , Souris transgéniques , Rayonnement ionisant , Récepteurs CCR4/déficit , Récepteurs CCR4/métabolisme , Transduction du signal/effets des radiations , Peau/anatomopathologie , Peau/effets des radiations
14.
PLoS One ; 16(3): e0247501, 2021.
Article de Anglais | MEDLINE | ID: mdl-33730087

RÉSUMÉ

Murine bone marrow (BM) chimeras are a versatile and valuable research tool in stem cell and immunology research. Engraftment of donor BM requires myeloablative conditioning of recipients. The most common method used for mice is ionizing radiation, and Cesium-137 gamma irradiators have been preferred. However, radioactive sources are being out-phased worldwide due to safety concerns, and are most commonly replaced by X-ray sources, creating a need to compare these sources regarding efficiency and potential side effects. Prior research has proven both methods capable of efficiently ablating BM cells and splenocytes in mice, but with moderate differences in resultant donor chimerism across tissues. Here, we compared Cesium-137 to 350 keV X-ray irradiation with respect to immune reconstitution, assaying complete, syngeneic BM chimeras and a mixed chimera model of autoimmune disease. Based on dose titration, we find that both gamma and X-ray irradiation can facilitate a near-complete donor chimerism. Mice subjected to 13 Gy Cesium-137 irradiation and reconstituted with syngeneic donor marrow were viable and displayed high donor chimerism, whereas X-ray irradiated mice all succumbed at 13 Gy. However, a similar degree of chimerism as that obtained following 13 Gy gamma irradiation could be achieved by 11 Gy X-ray irradiation, about 85% relative to the gamma dose. In the mixed chimera model of autoimmune disease, we found that a similar autoimmune phenotype could be achieved irrespective of irradiation source used. It is thus possible to compare data generated, regardless of the irradiation source, but every setup and application likely needs individual optimization.


Sujet(s)
Maladies auto-immunes/immunologie , Cellules de la moelle osseuse/effets des radiations , Transplantation de moelle osseuse/méthodes , Moelle osseuse/effets des radiations , Radio-isotopes du césium , Rayons gamma , Chimère post-radique/immunologie , Animaux , Modèles animaux de maladie humaine , Femelle , Maladie du greffon contre l'hôte/immunologie , Mâle , Souris , Souris de lignée C57BL , Irradiation corporelle totale , Rayons X
15.
Int J Radiat Biol ; 97(4): 517-528, 2021.
Article de Anglais | MEDLINE | ID: mdl-33591845

RÉSUMÉ

PURPOSE: To determine the early- and late-occurring damage in the bone marrow (BM) and peripheral blood cells of male CBA/Ca mice after exposure to 0, 0.1, 0.25, or 0.5 Gy of 1 GeV/n titanium (48Ti) ions (one type of space radiation). METHOD: We used the mouse in vivo blood-erythrocyte micronucleus (MN) assay for evaluating the cytogenetic effects of various doses of 1 GeV/n 48Ti ions. The MN assay was coupled with the characterization of epigenetic alterations (the levels of global 5-methylcytosine and 5-hydroxymethylcytosine) in DNA samples isolated from BM cells. These analyses were performed in samples collected at an early time-point (1 week) and a late time-point (6 months) post-irradiation. RESULTS: Our results showed that 48Ti ions induced genomic instability in exposed mice. Significant dose-dependent loss of global 5-hydroxymethylcytosine was found but there were no changes in global 5-methylcytosine levels. CONCLUSION: Since persistent genomic instability and loss of global 5-hydroxymethylcytosine are linked to cancer, our findings suggest that exposure to 48Ti ions may pose health risks.


Sujet(s)
Cellules de la moelle osseuse/effets des radiations , Titane/effets indésirables , Irradiation corporelle totale/effets indésirables , Animaux , Cellules de la moelle osseuse/métabolisme , Altération de l'ADN , Relation dose-effet des rayonnements , Rayons gamma/effets indésirables , Mâle , Souris , Souris de lignée CBA , Radio-isotopes/effets indésirables
16.
Radiat Res ; 195(3): 301-306, 2021 03 01.
Article de Anglais | MEDLINE | ID: mdl-33347573

RÉSUMÉ

Mouse models of radiation-induced thymic lymphoma are commonly used to study the biological effects of total-body irradiation (TBI) on the formation of hematologic malignancies. It is well documented that radiation-induced thymic lymphoma can be inhibited by protecting the bone marrow (BM) from irradiation; however, the mechanisms underlying this phenomenon are poorly understood. Here, we aimed to address this question by performing transplantation of BM cells from genetically engineered mice that have defects in tumor immunosurveillance or occupying different thymic niches. We found that BM cells from mice that have impaired tumor immunosurveillance, by deleting tumor necrosis factor alpha (TNFα), interferon gamma (IFNγ) or perforin-1 (PRF1), remained sufficient to suppress the formation of radiation-induced thymic lymphoma. On the other hand, BM cells from Rag2-/-; γc-/- mice and Rag2-/- mice, which have defects in occupying thymic niches beyond double negative (DN2) and DN3, respectively, failed to inhibit radiation-induced lymphomagenesis in the thymus. Taken together, based on our findings, we propose a model where unirradiated BM cells suppress radiation-induced lymphomagenesis in the thymus by competing with tumor-initiating cells for thymic niches beyond the DN3 stage.


Sujet(s)
Transplantation de moelle osseuse , Lymphomes/thérapie , Tumeurs radio-induites/thérapie , Tumeurs du thymus/thérapie , Animaux , Cellules de la moelle osseuse/effets des radiations , Modèles animaux de maladie humaine , Humains , Lymphomes/étiologie , Lymphomes/prévention et contrôle , Souris , Souris de lignée C57BL , Tumeurs radio-induites/prévention et contrôle , Tumeurs du thymus/étiologie , Tumeurs du thymus/prévention et contrôle , Irradiation corporelle totale/effets indésirables
17.
Radiat Res ; 195(2): 191-199, 2021 02 01.
Article de Anglais | MEDLINE | ID: mdl-33302291

RÉSUMÉ

Exposure to acute, high-dose, whole-body ionizing radiation results in bone marrow failure (hematopoietic acute radiation syndrome with resultant infection, bleeding, anemia, and increased risk of death). Sargramostim (yeast-derived rhu GM-CSF), a yeast-derived, molecularly cloned, hematopoietic growth factor and pleiotropic cytokine supports proliferation, differentiation, maturation and survival of cells of several myeloid lineages. We evaluated the efficacy of sargramostim in non-human primates (rhesus macaques) exposed to whole-body ionizing radiation at a 50-60% lethal dose. The primary end point was day 60 survival. Non-human primates received daily subcutaneous sargramostim (7 mcg/kg/day) or control. To reflect the anticipated setting of a nuclear or radiologic event, treatment began 48 h postirradiation, and non-human primates received only moderate supportive care (no whole blood transfusions or individualized antibiotics). Sargramostim significantly increased day 60 survival to 78% (95% confidence interval, 61-90%) vs. 42% (26-59%; P = 0.0018) in controls. Neutrophil, platelet and lymphocyte recovery rates were accelerated and infection rates decreased. Improved survival when sargramostim was started 48 h postirradiation, without use of intensive supportive care, suggests sargramostim may be effective in treating humans exposed to acute, high-dose whole-body, ionizing radiation in a scenario such as a mass casualty event.


Sujet(s)
Syndrome d'irradiation aigu/traitement médicamenteux , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Aplasies médullaires/traitement médicamenteux , Facteur de stimulation des colonies de granulocytes et de macrophages/pharmacologie , Syndrome d'irradiation aigu/génétique , Syndrome d'irradiation aigu/anatomopathologie , Animaux , Moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/effets des radiations , Aplasies médullaires/génétique , Aplasies médullaires/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des radiations , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des radiations , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des radiations , Facteur de stimulation des colonies de granulocytes , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Humains , Macaca mulatta/génétique , Mâle , Protéines recombinantes/pharmacologie , Irradiation corporelle totale/effets indésirables
18.
Sci Rep ; 10(1): 22211, 2020 12 17.
Article de Anglais | MEDLINE | ID: mdl-33335275

RÉSUMÉ

The only available option to treat radiation-induced hematopoietic syndrome is allogeneic hematopoietic cell transplantation, a therapy unavailable to many patients undergoing treatment for malignancy, which would also be infeasible in a radiological disaster. Stromal cells serve as critical components of the hematopoietic stem cell niche and are thought to protect hematopoietic cells under stress. Prior studies that have transplanted mesenchymal stromal cells (MSCs) without co-administration of a hematopoietic graft have shown underwhelming rescue of endogenous hematopoiesis and have delivered the cells within 24 h of radiation exposure. Herein, we examine the efficacy of a human bone marrow-derived MSC therapy delivered at 3 h or 30 h in ameliorating radiation-induced hematopoietic syndrome and show that pancytopenia persists despite MSC therapy. Animals exposed to radiation had poorer survival and experienced loss of leukocytes, platelets, and red blood cells. Importantly, mice that received a therapeutic dose of MSCs were significantly less likely to die but experienced equivalent collapse of the hematopoietic system. The cause of the improved survival was unclear, as complete blood counts, splenic and marrow cellularity, numbers and function of hematopoietic stem and progenitor cells, and frequency of niche cells were not significantly improved by MSC therapy. Moreover, human MSCs were not detected in the bone marrow. MSC therapy reduced crypt dropout in the small intestine and promoted elevated expression of growth factors with established roles in gut development and regeneration, including PDGF-A, IGFBP-3, IGFBP-2, and IGF-1. We conclude that MSC therapy improves survival not through overt hematopoietic rescue but by positive impact on other radiosensitive tissues, such as the intestinal mucosa. Collectively, these data reveal that MSCs could be an effective countermeasure in cancer patients and victims of nuclear accidents but that MSCs alone do not significantly accelerate or contribute to recovery of the blood system.


Sujet(s)
Hématopoïèse/effets des radiations , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/métabolisme , Lésions radiques/mortalité , Lésions radiques/thérapie , Animaux , Biopsie , Moelle osseuse/métabolisme , Moelle osseuse/anatomopathologie , Moelle osseuse/effets des radiations , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/effets des radiations , Modèles animaux de maladie humaine , Femelle , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/effets des radiations , Humains , Immunophénotypage , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/effets des radiations , Mâle , Cellules souches mésenchymateuses/cytologie , Pancytopénie/étiologie , Pancytopénie/métabolisme , Pancytopénie/anatomopathologie , Pronostic , Lésions radiques/anatomopathologie , Radiothérapie/effets indésirables , Résultat thérapeutique
19.
Probl Radiac Med Radiobiol ; 25: 300-308, 2020 Dec.
Article de Anglais, Ukrainien | MEDLINE | ID: mdl-33361842

RÉSUMÉ

OBJECTIVE: To perform comparative analysis of the characteristics of population functioning process of mice bonemarrow colony-forming units after their prolonged irradiation in lethal and non-lethal doses with equal dose rateintensity with the aid of mathematical model. MATERIALS AND METHODS: Assigned task is solved by means of mathematical model of alterations in the number ofbone marrow colony-forming units after continuous irradiation, described in previous works, with the use of experimental results of K. S. Chertkov works (1972, 1973). Mathematical model is developed basing on the hematopoiesisscheme introduced by I. L. Chertkov (1984, 1991). RESULTS AND CONCLUSIONS: By applying original mathematical model, new scheme of hematopoiesis [6], with theuse of experimental results of γ-irradiation influence in the doses of 4 Gy and 8 Gy with the dose rate intensity of0.0028 Gy/min on the number of mice bone marrow colony-forming units, as well as experimental data concerningthe processes of their number recovery, obtained from literature references, we determined the parameters, whichcharacterize hematopoietic system reaction on the different stages of recovery processes of mice bone marrowcolony-forming units after the termination of ionizing radiation action; comparative analysis of obtained resultswas performed.


Sujet(s)
Cellules de la moelle osseuse/effets des radiations , Moelle osseuse/effets des radiations , Hématopoïèse/effets des radiations , Cellules souches hématopoïétiques/effets des radiations , Modèles théoriques , Lésions radiques/anatomopathologie , Animaux , Moelle osseuse/anatomopathologie , Cellules de la moelle osseuse/anatomopathologie , Chimère , Test clonogénique , Relation dose-effet des rayonnements , Femelle , Rayons gamma/effets indésirables , Cellules souches hématopoïétiques/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris de lignée CBA , Lésions radiques/mortalité , Analyse de survie , Irradiation corporelle totale
20.
Probl Radiac Med Radiobiol ; 25: 443-455, 2020 Dec.
Article de Anglais, Ukrainien | MEDLINE | ID: mdl-33361853

RÉSUMÉ

OBJECTIVE: Describe and characterize the peculiarities of the chronic myeloid leukemia (CML) course and responseto treatment in patients irradiated as a result of the Chornobyl nuclear power plant (ChNPP) accident based on theassessment of clinical-laboratory and clinical parameters. MATERIALS AND METHODS: The CML patients (n = 33) exposed to ionizing radiation as a result of the ChNPP accidentwere enrolled. The comparison group consisted of CML patients (n = 725) with no history of radiation exposure. Allpatients were in the chronic phase of the disease. Clinical, hematological and molecular genetic research methodswere applied. RESULTS: Patients exposed to ionizing radiation as a result of the ChNPP accident had no differences in CML manifestation, as well as in classical genetic markers at the onset of the disease compared with patients with no historyof radiation exposure. Reduction of tumor clone on imatinib therapy was significantly less effective in the patientsexposed to ionizing radiation than in cases of no history of radiation exposure. Cases of primary resistance were statistically significantly prevalent in the ChNPP accident consequences clean-up workers while in the residents ofradiologically contaminated areas a statistically significant increase in probability of loss of complete cytogeneticresponse (development of secondary resistance) to imatinib therapy was found. An association was found betweenthe radiation exposure and probability of loss of complete cytogenetic response to imatinib therapy in this group ofpatients. CONCLUSION: The radiation exposure in the history even many years before the onset of CML is an unfavorable exogenous factor responsible for the development of resistance to imatinib therapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Accident nucléaire de Tchernobyl , Protéines de fusion bcr-abl/génétique , Leucémie myéloïde chronique BCR-ABL positive/génétique , Exposition aux rayonnements/effets indésirables , Lésions radiques/génétique , Sujet âgé , Polluants atmosphériques radioactifs/effets indésirables , Cellules de la moelle osseuse/immunologie , Cellules de la moelle osseuse/anatomopathologie , Cellules de la moelle osseuse/effets des radiations , Chromosomes humains de la paire 22 , Chromosomes humains de la paire 9 , Résistance aux médicaments antinéoplasiques/génétique , Intervenants d'urgence , Femelle , Contamination radioactive des aliments , Expression des gènes , Humains , Mésilate d'imatinib/usage thérapeutique , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/étiologie , Leucémie myéloïde chronique BCR-ABL positive/mortalité , Mâle , Adulte d'âge moyen , Pronostic , Lésions radiques/traitement médicamenteux , Lésions radiques/étiologie , Lésions radiques/mortalité , Rayonnement ionisant , Polluants radioactifs du sol/effets indésirables , Analyse de survie , Translocation génétique , Ukraine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...