Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 12(1): 1-15, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32985332

ABSTRACT

Every year, millions of people around the world benefit from radiation therapy to treat cancers localized in the pelvic area. Damage to healthy tissue in the radiation field can cause undesirable toxic effects leading to gastrointestinal complications called pelvic radiation disease. A change in the composition and/or function of the microbiota could contribute to radiation-induced gastrointestinal toxicity. In this study, we tested the prophylactic effect of a new generation of probiotic like Faecalibacterium prausnitzii (F. prausnitzii) on acute radiation-induced colonic lesions. Experiments were carried out in a preclinical model of pelvic radiation disease. Rats were locally irradiated at 29 Gray in the colon resulting in colonic epithelial barrier rupture. Three days before the irradiation and up to 3 d after the irradiation, the F. prausnitzii A2-165 strain was administered daily (intragastrically) to test its putative protective effects. Results showed that prophylactic F. prausnitzii treatment limits radiation-induced para-cellular hyperpermeability, as well as the infiltration of neutrophils (MPO+ cells) in the colonic mucosa. Moreover, F. prausnitzii treatment reduced the severity of the morphological change of crypts, but also preserved the pool of Sox-9+ stem/progenitor cells, the proliferating epithelial PCNA+ crypt cells and the Dclk1+/IL-25+ differentiated epithelial tuft cells. The benefit of F. prausnitzii was associated with increased production of IL-18 by colonic crypt epithelial cells. Thus, F. prausnitzii treatment protected the epithelial colonic barrier from colorectal irradiation. New-generation probiotics may be promising prophylactic treatments to reduce acute side effects in patients treated with radiation therapy and may improve their quality of life.


Subject(s)
Colon/radiation effects , Faecalibacterium prausnitzii , Intestinal Mucosa/radiation effects , Probiotics , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control , Animals , Cell Proliferation , Colon/immunology , Colon/pathology , Colon/physiopathology , Gastrointestinal Microbiome , Interleukin-18/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Macrophages/physiology , Male , Neutrophils/physiology , Pelvis , Permeability , Radiation Injuries, Experimental/immunology , Rats , Rats, Sprague-Dawley , Rectum/radiation effects , Stem Cells/physiology
2.
Stem Cell Res Ther ; 11(1): 371, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32854778

ABSTRACT

BACKGROUND: Human exposure to high doses of radiation resulting in acute radiation syndrome and death can rapidly escalate to a mass casualty catastrophe in the event of nuclear accidents or terrorism. The primary reason is that there is presently no effective treatment option, especially for radiation-induced gastrointestinal syndrome. This syndrome results from disruption of mucosal barrier integrity leading to severe dehydration, blood loss, and sepsis. In this study, we tested whether extracellular vesicles derived from mesenchymal stromal cells (MSC) could reduce radiation-related mucosal barrier damage and reduce radiation-induced animal mortality. METHODS: Human MSC-derived extracellular vesicles were intravenously administered to NUDE mice, 3, 24, and 48 h after lethal whole-body irradiation (10 Gy). Integrity of the small intestine epithelial barrier was assessed by morphologic analysis, immunostaining for tight junction protein (claudin-3), and in vivo permeability to 4 kDa FITC-labeled dextran. Renewal of the small intestinal epithelium was determined by quantifying epithelial cell apoptosis (TUNEL staining) and proliferation (Ki67 immunostaining). Statistical analyses were performed using one-way ANOVA followed by a Tukey test. Statistical analyses of mouse survival were performed using Kaplan-Meier and Cox methods. RESULTS: We demonstrated that MSC-derived extracellular vesicle treatment reduced by 85% the instantaneous mortality risk in mice subjected to 10 Gy whole-body irradiation and so increased their survival time. This effect could be attributed to the efficacy of MSC-derived extracellular vesicles in reducing mucosal barrier disruption. We showed that the MSC-derived extracellular vesicles improved the renewal of the small intestinal epithelium by stimulating proliferation and inhibiting apoptosis of the epithelial crypt cells. The MSC-derived extracellular vesicles also reduced radiation-induced mucosal permeability as evidenced by the preservation of claudin-3 immunostaining at the tight junctions of the epithelium. CONCLUSIONS: MSC-derived extracellular vesicles promote epithelial repair and regeneration and preserve structural integrity of the intestinal epithelium in mice exposed to radiation-induced gastrointestinal toxicity. Our results suggest that the administration of MSC-derived extracellular vesicles could be an effective therapy for limiting acute radiation syndrome.


Subject(s)
Acute Radiation Syndrome , Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Intestinal Mucosa , Intestines , Mice , Mice, Nude
3.
Biomaterials ; 115: 40-52, 2017 01.
Article in English | MEDLINE | ID: mdl-27886554

ABSTRACT

Healthy tissues surrounding abdomino-pelvic tumours can be impaired by radiotherapy, leading to chronic gastrointestinal complications with substantial mortality. Adipose-derived Mesenchymal Stromal Cells (Ad-MSCs) represent a promising strategy to reduce intestinal lesions. However, systemic administration of Ad-MSCs results in low cell engraftment within the injured tissue. Biomaterials, able to encapsulate and withstand Ad-MSCs, can overcome these limitations. A silanized hydroxypropylmethyl cellulose (Si-HPMC) hydrogel has been designed and characterized for injectable cell delivery using the operative catheter of a colonoscope. We demonstrated that hydrogel loaded-Ad-MSCs were viable, able to secrete trophic factors and responsive to the inflammatory environment. In a rat model of radiation-induced severe colonic damage, Ad-MSC + Si-HPMC improve colonic epithelial structure and hyperpermeability compared with Ad-MSCs injected intravenously or locally. This therapeutic benefit is associated with greater engraftment of Si-HPMC-embedded Ad-MSCs in the irradiated colonic mucosa. Moreover, macrophage infiltration near the injection site was less pronounced when Ad-MSCs were embedded in the hydrogel. Si-HPMC induces modulation of chemoattractant secretion by Ad-MSCs that could contribute to the decrease in macrophage infiltrate. Si-HPMC is suitable for cell delivery by colonoscopy and induces protection of Ad-MSCs in the tissue potentiating their therapeutic effect and could be proposed to patients suffering from colon diseases.


Subject(s)
Colonic Diseases/pathology , Colonic Diseases/therapy , Hydrogels/chemistry , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Radiation Injuries/pathology , Radiation Injuries/therapy , Animals , Biocompatible Materials/chemistry , Cells, Cultured , Colonic Diseases/etiology , Male , Radiation Injuries/etiology , Radiotherapy, Conformal/adverse effects , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Tissue Scaffolds , Treatment Outcome
4.
Cell Transplant ; 25(10): 1723-1746, 2016 10.
Article in English | MEDLINE | ID: mdl-27197023

ABSTRACT

Ionizing radiation is effective to treat malignant pelvic cancers, but the toxicity to surrounding healthy tissue remains a substantial limitation. Early and late side effects not only limit the escalation of the radiation dose to the tumor but may also be life-threatening in some patients. Numerous preclinical studies determined specific mechanisms induced after irradiation in different compartments of the intestine. This review outlines the complexity of the pathogenesis, highlighting the roles of the epithelial barrier in the vascular network, and the inflammatory microenvironment, which together lead to chronic fibrosis. Despite the large number of pharmacological molecules available, the studies presented in this review provide encouraging proof of concept regarding the use of mesenchymal stromal cell (MSC) therapy to treat radiation-induced intestinal damage. The therapeutic efficacy of MSCs has been demonstrated in animal models and in patients, but an enormous number of cells and multiple injections are needed due to their poor engraftment capacity. Moreover, it has been observed that although MSCs have pleiotropic effects, some intestinal compartments are less restored after a high dose of irradiation. Future research should seek to optimize the efficacy of the injected cells, particularly with regard to extending their life span in the irradiated tissue. Moreover, improving the host microenvironment, combining MSCs with other specific regenerative cells, or introducing new tissue engineering strategies could be tested as methods to treat the severe side effects of pelvic radiotherapy.


Subject(s)
Cell- and Tissue-Based Therapy , Radiation Injuries/pathology , Tissue Engineering , Animals , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology , Intestines/radiation effects , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Microbiota , Oxidative Stress/radiation effects , Radiation Injuries/therapy , Vascular Endothelial Growth Factor A/metabolism
5.
J Pathol ; 237(4): 435-46, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26177977

ABSTRACT

Radiation proctitis is an insidious disease associated with substantial morbidity and mortality. It may develop following the treatment of several cancers by radiotherapy when normal colorectal tissues are present in the irradiation field. There is no unified approach for the assessment and treatment of this disease, partly due to insufficient knowledge about the mechanism involved in the development of radiation proctitis. However, unresolved inflammation is hypothesized to have an important role in late side effects. This study aimed to analyse the involvement of specific immunity in colorectal damage developing after localized irradiation, and evaluate the benefit of immunomodulatory mesenchymal stromal cells isolated from adipose tissue (Ad-MSCs) for reduction of late side effects. Our experimental model of colorectal irradiation induced severe colonic mucosal damage and fibrosis that was associated with T-cell infiltration. Immune cell activation was investigated; adoptive transfer of T cells in nude rats showed stronger colonization by T cells isolated from irradiated rats. The predominant role of T cells in late radiation-induced damage and regeneration processes was highlighted by in vivo depletion experiments. Treatments using Ad-MSCs reduced T-cell infiltration in the colon and reduced established colonic damage as measured by histological score, functional circular muscle contractibility, and collagen deposition. Here, we have demonstrated for the first time the predominance of the TH17 population compared to TH1 and TH2 in radiation-induced bowel disease, and that this is reduced after Ad-MSC treatment. Additionally, we demonstrated in vitro that IL17 acts directly on colonic smooth muscle cells to induce expression of pro-inflammatory genes that could participate in the development of radiation-induced injury. Our data demonstrate that the TH17 population is specifically induced during development of radiation-induced side effects in the colon. Moreover, Ad-MSC treatment modulates the TH17 population and reduces the extracellular matrix remodelling process induced following irradiation.


Subject(s)
Intestines/radiation effects , Lymphocyte Activation/immunology , Mesenchymal Stem Cell Transplantation/methods , Radiation Injuries, Experimental/immunology , Th17 Cells/immunology , Adipose Tissue/cytology , Adoptive Transfer , Animals , Disease Models, Animal , Intestines/immunology , Intestines/pathology , Male , Mesenchymal Stem Cells , Radiation Injuries, Experimental/pathology , Rats , Rats, Nude , Rats, Sprague-Dawley
6.
Pain ; 156(8): 1465-1476, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25887464

ABSTRACT

Each year, millions of people worldwide are treated for primary or recurrent pelvic malignancies, involving radiotherapy in almost 50% of cases. Delayed development of visceral complications after radiotherapy is recognized in cancer survivors. Therapeutic doses of radiation may lead to the damage of healthy tissue around the tumor and abdominal pain. Because of the lack of experimental models, the underlying mechanisms of radiation-induced long-lasting visceral pain are still unknown. This makes managing radiation-induced pain difficult, and the therapeutic strategies proposed are mostly inefficient. The aim of our study was to develop an animal model of radiation-induced visceral hypersensitivity to (1) analyze some cellular and molecular mechanisms involved and (2) to test a therapeutic strategy using mesenchymal stromal cells (MSCs). Using a single 27-Grays colorectal irradiation in rats, we showed that such exposure induces a persistent visceral allodynia that is associated with an increased spinal sensitization (enhanced p-ERK neurons), colonic neuroplasticity (as increased density of substance P nerve fibers), and colonic mast cell hyperplasia and hypertrophy. Mast cell stabilization by ketotifen provided evidence of their functional involvement in radiation-induced allodynia. Finally, intravenous injection of 1.5 million MSCs, 4 weeks after irradiation, induced a time-dependent reversion of the visceral allodynia and a reduction of the number of anatomical interactions between mast cells and PGP9.5+ nerve fibers. Moreover, unlike ketotifen, MSC treatment has the key advantage to limit radiation-induced colonic ulceration. This work provides new insights into the potential use of MSCs as cellular therapy in the treatment of pelvic radiation disease.


Subject(s)
Abdominal Pain/therapy , Colon/radiation effects , Hyperalgesia/therapy , Mesenchymal Stem Cell Transplantation/methods , Radiation Injuries, Experimental/complications , Abdominal Pain/etiology , Abdominal Pain/metabolism , Animals , Colon/pathology , Disease Models, Animal , Hyperalgesia/metabolism , Hyperplasia/pathology , Hypertrophy/pathology , Male , Mast Cells/metabolism , Mesenchymal Stem Cells/metabolism , Neuronal Plasticity , Protein Tyrosine Phosphatases/metabolism , Rats , Treatment Outcome
7.
PLoS One ; 8(7): e70170, 2013.
Article in English | MEDLINE | ID: mdl-23922953

ABSTRACT

Patients who undergo pelvic radiotherapy may develop severe and chronic complications resulting from gastrointestinal alterations. The lack of curative treatment highlights the importance of novel and effective therapeutic strategies. We thus tested the therapeutic benefit of mesenchymal stem cells (MSC) treatment and proposed molecular mechanisms of action. MSC efficacy was tested in an experimental model of radiation-induced severe colonic ulceration histologically similar to that observed in patients. In this model, MSC from bone marrow were administered intravenously, immediately or three weeks (established lesions) after irradiation. MSC therapy reduces radiation-induced colonic ulceration and increases animal survival. MSC treatment induces therapeutic efficacy whatever the time of cell infusion. Infused-MSC engraft in the colon but also increase endogenous MSC mobilization in blood that have lasting benefits over time. In vitro analysis demonstrates that the MSC effect is mediated by paracrine mechanisms through the non-canonical WNT (Wingless integration site) pathway. In irradiated rat colons, MSC treatment increases the expression of the non-canonical WNT4 ligand by epithelial cells. The epithelial regenerative process is improved after MSC injection by stimulation of colonic epithelial cells positive for SOX9 (SRY-box containing gene 9) progenitor/stem cell markers. This study demonstrates that MSC treatment induces stimulation of endogenous host progenitor cells to improve the regenerative process and constitutes an initial approach to arguing in favor of the use of MSC to limit/reduce colorectal damage induced by radiation.


Subject(s)
Colon/metabolism , Intestinal Mucosa/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Regeneration , Animals , Cell Movement , Cell Proliferation , Cell- and Tissue-Based Therapy , Colon/pathology , Colon/radiation effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Male , Rats , Ulcer/etiology , Wnt Signaling Pathway , Wnt4 Protein/metabolism
8.
Ann Hematol ; 86(1): 1-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17043780

ABSTRACT

It has been suggested that human mesenchymal stem cells (hMSC) could be used to repair numerous injured tissues. We have studied the potential use of hMSC to limit radiation-induced skin lesions. Immunodeficient NOD/SCID mice were locally irradiated to the leg (30 Gy, dose rate 2.7 Gy/min) using a (60)Co source to induce a severe skin lesion. Cultured bone marrow hMSC were delivered intravenously to the mice. The irradiated skin samples were studied for the presence of the human cells, the severity of the lesions and the healing process. Macroscopic analysis and histology results showed that the lesions were evolving to a less severe degree of radiation dermatitis after hMSC transplant when compared to irradiated non-transplanted controls. Clinical scores for the studied skin parameters of treated mice were significantly improved. A faster healing was observed when compared to untreated mouse. Immunohistology and polymerase chain reaction analysis provided evidence that the human cells were found in the irradiated area. These results suggest a possible use of hMSC for the treatment of the early phase of the cutaneous radiation syndrome. A successful transplant of stem cells and subsequent reduction in radiation-induced complication may open the road to completely new strategies in cutaneous radiation syndrome therapy.


Subject(s)
Mesenchymal Stem Cell Transplantation , Radiodermatitis/therapy , Transplantation, Heterologous , Animals , Bone Marrow Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Radiodermatitis/pathology , Syndrome , Wound Healing
9.
Adv Exp Med Biol ; 585: 19-30, 2006.
Article in English | MEDLINE | ID: mdl-17120774

ABSTRACT

Patients who undergo pelvic or abdominal radiotherapy may develop side effects that can be life threatening. Tissue complications caused by radiation-induced stem cell depletion may result in structural and functional alterations of the gastrointestinal (GI) tract. Stem cell therapy using mesenchymal stem cells (MSC) is a promising approach for replenishment of the depleted stem cell compartment during radiotherapy. There is little information on the therapeutic potential of MSC in injured-GI tract following radiation exposure. In this study, we addressed the ability of MSC to support the structural regeneration of the small intestine after abdominal irradiation. We isolated MSC from human bone marrow and human mesenchymal stem cells (hMSC) were transplanted into immunotolerent NOD/SCID mice with a dose of 5.10(6) cells via the systemic route. Using a model of radiation-induced intestinal injury, we studied the link between damage, hMSC engraftment and the capacity of hMSC to sustain structural recovery. Tissue injury was assessed by histological analysis. hMSC engraftment in tissues was quantified by PCR assay. Following abdominal irradiation, the histological analysis of small intestinal structure confirms the presence of partial and transient (three days) mucosal atrophy. PCR analysis evidences a low but significant hMSC implantation in small intestine (0.17%) but also at all the sites of local irradiation (kidney, stomach and spleen). Finally, in presence of hMSC, the small intestinal structure is already recovered at three days after abdominal radiation exposure. We show a structural recovery accompanied by an increase of small intestinal villus height, three and fifteen days following abdominal radiation exposure. In this study, we show that radiation-induced small intestinal injury may play a role in the recruitment of MSC for the improvement of tissue recovery. This work supports, the use of MSC infusion to repair damaged GI tract in patients subjected to radiotherapy. MSC therapy to avoid extended intestinal crypt sterilization is a promising approach to diminish healthy tissue alterations during the course of pelvic radiotherapy.


Subject(s)
Epithelial Cells/cytology , Intestine, Small/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Radiation Injuries, Experimental , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Epithelial Cells/radiation effects , Humans , Intestine, Small/pathology , Intestine, Small/radiation effects , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Inbred NOD , Mice, SCID , Regeneration , Stem Cell Transplantation/methods
10.
Stem Cells ; 24(4): 1020-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16339642

ABSTRACT

Mesenchymal stem cells (MSCs) have been shown to migrate to various tissues. There is little information on the fate and potential therapeutic efficacy of the reinfusion of MSCs following total body irradiation (TBI). We addressed this question using human MSC (hMSCs) infused to nonobese diabetic/ severe combined immunodeficient (NOD/SCID) mice submitted to TBI. Further, we tested the impact of additional local irradiation (ALI) superimposed to TBI, as a model of accidental irradiation. NOD/SCID mice were transplanted with hM-SCs. Group 1 was not irradiated before receiving hMSC infusion. Group 2 received only TBI at a dose of 3.5 Gy, group 3 received local irradiation to the abdomen at a dose of 4.5 Gy in addition to TBI, and group 4 received local irradiation to the leg at 26.5 Gy in addition to TBI. Fifteen days after irradiation, quantitative and spatial distribution of the hMSCs were studied. Histological analysis of mouse tissues confirmed the presence of radio-induced lesions in the irradiated fields. Following their infusion into nonirradiated animals, hMSCs homed at a very low level to various tissues (lung, bone marrow, and muscles) and no significant engraftment was found in other organs. TBI induced an increase of engraftment levels of hMSCs in the brain, heart, bone marrow, and muscles. Abdominal irradiation (AI) as compared with leg irradiation (LI) increased hMSC engraftment in the exposed area (the gut, liver, and spleen). Hind LI as compared with AI increased hMSC engraftment in the exposed area (skin, quadriceps, and muscles). An increase of hMSC engraftment in organs outside the fields of the ALI was also observed. Conversely, following LI, hMSC engraftment was increased in the brain as compared with AI. This study shows that engraftment of hMSCs in NOD/ SCID mice with significantly increased in response to tissue injuries following TBI with or without ALI. ALI induced an increase of the level of engraftment at sites outside the local irradiation field, thus suggesting a distant (abscopal) effect of radiation damage. This work supports the use of MSCs to repair damaged normal tissues following accidental irradiation and possibly in patients submitted to radiotherapy.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/radiation effects , Animals , Cell Movement/radiation effects , Gene Expression , Globins/genetics , Graft Survival/genetics , Graft Survival/radiation effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Organ Specificity , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/therapy , Transplantation, Heterologous , Whole-Body Irradiation , beta 2-Microglobulin/metabolism
11.
Oncogene ; 23(52): 8497-508, 2004 Nov 04.
Article in English | MEDLINE | ID: mdl-15361846

ABSTRACT

We investigated the role of tumor suppressor p53 and Fas (CD95/APO-1), a member of the tumor necrosis factor receptor family, in neural progenitors response to gamma-irradiation exposure. Telencephalic cells were obtained from wild-type C57Bl/6, or p53-/- or fas-/-, 15-day-old mouse embryos. They were cultured in conditions allowing neural progenitors to form proliferating clusters (neurospheres). A 2 Gy gamma-irradiation induced a G1 cell cycle arrest and triggered apoptosis in wild-type neural progenitor cultures in correlation with an enhanced expression of p53 and of its downstream target p21(WAF1), both of them acquiring a nuclear localization. These effects did not occur in p53-/- neural progenitors demonstrating the central role played by p53 in their response to ionizing radiation. Furthermore, the monoclonal antibody Jo2 directed against Fas induced apoptosis of wild type but not of fas-/- neural progenitors, indicating the existence of a functional Fas signaling pathway in neural progenitors. Ionizing radiation induced an increase of Fas membrane expression related to a p53-dependent increase of fas mRNA expression in wild-type neural progenitors. Moreover, fas-/- neural progenitors exhibited delayed radiation-induced apoptosis compared to wild-type cells. Therefore, these findings establish a role for Fas/CD95 related to p53 in the response of neural progenitors to gamma-radiation exposure. Similar mechanisms could be triggered in neural progenitors in case of different stresses during brain development or in the course of various diseases affecting the adult brain.


Subject(s)
Gamma Rays , Receptors, Tumor Necrosis Factor/metabolism , Stem Cells/radiation effects , Tumor Suppressor Protein p53/metabolism , fas Receptor/metabolism , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Cell Cycle/physiology , Cell Cycle/radiation effects , Mice , Signal Transduction/physiology , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...