Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sensors (Basel) ; 22(14)2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35890977

ABSTRACT

Unmanned Aerial Vehicles (UAVs) are often studied as tools to perform data collection from Wireless Sensor Networks (WSNs). Path planning is a fundamental aspect of this endeavor. Works in the current literature assume that data are always ready to be retrieved when the UAV passes. This operational model is quite rigid and does not allow for the integration of the UAV as a computational object playing an active role in the network. In fact, the UAV could begin the computation on a first visit and retrieve the data later. Potentially, the UAV could orchestrate the distributed computation to improve its performance, change its parameters, and even upload new applications to the sensor network. In this paper, we analyze a scenario where a UAV plays an active role in the operation of multiple sensor networks by visiting different node clusters to initiate distributed computation and collect the final outcomes. The experimental results validate the effectiveness of the proposed method in optimizing total flight time, Average Age of Information, Average cluster computation end time, and Average data collection time compared to prevalent approaches to UAV path-planning that are adapted to the purpose.

2.
Contrast Media Mol Imaging ; 2018: 3064751, 2018.
Article in English | MEDLINE | ID: mdl-30154685

ABSTRACT

The bile acid analogue [18F]LCATD (LithoCholic Acid Triazole Derivative) is transported in vitro by hepatic uptake transporters such as OATP1B1 and NTCP and efflux transporter BSEP. In this in vivo "proof of principle" study, we tested if [18F]LCATD may be used to evaluate drug-drug interactions (DDIs) caused by inhibition of liver transporters. Hepatic clearance of [18F]LCATD in rats was significantly modified upon coadministration of rifamycin SV or sodium fusidate, which are known to inhibit clinically relevant uptake transporters (OATP1B1, NTCP) and canalicular hepatic transporters (BSEP) in humans. Treatment with rifamycin SV (total dose 62.5 mg·Kg-1) reduced the maximum radioactivity of [18F]LCATD recorded in the liver from 14.2 ± 0.8% to 10.2 ± 0.9% and delayed t_max by 90 seconds relative to control rats. AUCliver 0-5 min, AUCbile 0-10 min and hepatic uptake clearance CLuptake,in vivo of rifamycin SV treated rats were significantly reduced, whereas AUCliver 0-30 min was higher than in control rats. Administration of sodium fusidate (30 mg·Kg-1) inhibited the liver uptake of [18F]LCATD, although to a lesser extent, reducing the maximum radioactivity in the liver to 11.5 ± 0.3%. These preliminary results indicate that [18F]LCATD may be a good candidate for future applications as an investigational tracer to evaluate altered hepatobiliary excretion as a result of drug-induced inhibition of hepatic transporters.


Subject(s)
Drug Interactions , Fluorine Radioisotopes/chemistry , Liver/metabolism , Membrane Transport Proteins/metabolism , Positron-Emission Tomography , Triazoles/chemistry , Animals , Arteries/metabolism , Bile Ducts/metabolism , Female , Fluorine Radioisotopes/blood , Fluorine Radioisotopes/pharmacokinetics , Fusidic Acid/pharmacology , Kinetics , Organ Specificity , Rats, Sprague-Dawley , Rifamycins/pharmacology , Tissue Distribution , Triazoles/blood , Triazoles/pharmacokinetics
3.
Toxicol Lett ; 276: 138-143, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28495613

ABSTRACT

In a National Toxicology Program (NTP) bioassay, inhalation of tetrahydrofuran (THF) induced liver tumors in female B6C3F1 mice but not in male mice or rats of either sex. Since THF is not genotoxic, the NTP concluded this carcinogenic activity was likely mediated via non-genotoxic modes of action (MOA). Based on evidence that THF and phenobarbital share a similar MOA, female Car/Pxr knock-out mice were orally exposed to THF to evaluate the potential role of CAR activation in the MOA for THF-induced liver tumors. Because data from this oral study with Car/Pxr knock-out mice (C57Bl/6) and the inhalation studies with wild type mice (B6C3F1) reported by NTP and others were derived from different strains, oral studies with wild type B6C3F1 and C57Bl/6 mice were conducted to ensure THF responses in both strains were comparable. As seen in inhalation studies with THF, oral exposure of wild type female mice to a maximum tolerated dose of THF increased total P450 content, CAR-related P450 activities, and hepatocyte proliferation; these effects were not observed in Car/Pxr knock-out female mice. This finding supports the hypothesis THF-induced carcinogenicity is likely mediated via CAR activation that has limited, if any, relevance to humans.


Subject(s)
Carcinogens/toxicity , Furans/toxicity , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Administration, Oral , Animals , Carcinogens/administration & dosage , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Female , Furans/administration & dosage , Genotype , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Maximum Tolerated Dose , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/deficiency , Receptors, Steroid/genetics , Risk Assessment , Species Specificity
4.
Bioorg Med Chem ; 25(3): 963-976, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011201

ABSTRACT

With the aim of identifying a fluorinated bile acid derivative that could be used as [18F]-labeled Positron Emission Tomography (PET) tracer for imaging the in vivo functioning of liver transporter proteins, and particularly of OATP1B1, three fluorinated bile acid triazole derivatives of cholic, deoxycholic and lithocholic acid (CATD, DCATD and LCATD 4a-c, respectively) were synthesized and labeled with tritium. In vitro transport properties were studied with cell-based assays to identify the best substrate for OATP1B1. In addition, the lead compound, LCATD (4c), was tested as a substrate of other liver uptake transporters OATP1B3, NTCP and efflux transporter BSEP to evaluate its specificity of liver transport. The results suggest that 4c is a good substrate of OATP1B1 and NTCP, whereas it is a poor substrate of OATP1B3. The efflux transporter BSEP also appears to be involved in the excretion of 4c from hepatocytes. The automated radiosynthesis of [18F]-4c was accomplished in a multi-GBq scale and a pilot imaging experiment in a wild type rat was performed after i.v. administration to assess the biodistribution and clearance of the tracer. PET imaging revealed that radioactivity was primarily located in the liver (tmax=75s) and cleared exclusively through the bile, thus allowing to image the hepatobiliary excretion of bile acids in the animal model. These findings suggest that [18F]-LCATD 4c is a promising PET probe for the evaluation of hepatic transporters OATP1B1, NTCP and BSEP activity with potential for studying drug-drug interactions and drug-induced toxicity involving these transporters.


Subject(s)
Bile Acids and Salts/chemistry , Drug Design , Liver/metabolism , Positron-Emission Tomography , Animals , Bile Acids and Salts/chemical synthesis , Biological Transport , Female , Halogenation , Molecular Structure , Radioactive Tracers , Rats, Sprague-Dawley
5.
Ann Rheum Dis ; 75(7): 1386-91, 2016 07.
Article in English | MEDLINE | ID: mdl-26275430

ABSTRACT

BACKGROUND: Myeloid inhibitory C-type lectin-like receptor (MICL, Clec12A) is a C-type lectin receptor (CLR) expressed predominantly by myeloid cells. Previous studies have suggested that MICL is involved in controlling inflammation. OBJECTIVE: To determine the role of this CLR in inflammatory pathology using Clec12A(-/-) mice. METHODS: Clec12A(-/-) mice were generated commercially and primarily characterised using the collagen antibody-induced arthritis (CAIA) model. Mechanisms and progress of disease were characterised by clinical scoring, histology, flow cytometry, irradiation bone-marrow chimera generation, administration of blocking antibodies and in vivo imaging. Characterisation of MICL in patients with rheumatoid arthritis (RA) was determined by immunohistochemistry and single nucleotide polymorphism analysis. Anti-MICL antibodies were detected in patient serum by ELISA and dot-blot analysis. RESULTS: MICL-deficient animals did not present with pan-immune dysfunction, but exhibited markedly exacerbated inflammation during CAIA, owing to the inappropriate activation of myeloid cells. Polymorphisms of MICL were not associated with disease in patients with RA, but this CLR was the target of autoantibodies in a subset of patients with RA. In wild-type mice the administration of such antibodies recapitulated the Clec12A(-/-) phenotype. CONCLUSIONS: MICL plays an essential role in regulating inflammation during arthritis and is an autoantigen in a subset of patients with RA. These data suggest an entirely new mechanism underlying RA pathogenesis, whereby the threshold of myeloid cell activation can be modulated by autoantibodies that bind to cell membrane-expressed inhibitory receptors.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Lectins, C-Type/physiology , Receptors, Mitogen/physiology , Animals , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/pathology , Autoantibodies/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Lectins, C-Type/deficiency , Lectins, C-Type/immunology , Mice , Myeloid Cells/metabolism , Polymorphism, Genetic , Receptors, Mitogen/deficiency , Receptors, Mitogen/immunology , Synovial Membrane/pathology
7.
Arthritis Rheum ; 63(5): 1289-300, 2011 May.
Article in English | MEDLINE | ID: mdl-21538315

ABSTRACT

OBJECTIVE: We previously reported that human synovium contains cells that, after culture expansion, display properties of mesenchymal stem cells (MSCs). The objective of this study was to identify MSCs in native synovium in vivo. METHODS: To identify stem cells in the synovium in vivo, a double nucleoside analog cell-labeling scheme was used in a mouse model of joint-surface injury. For labeling of slow-cycling cells, mice received iododeoxyuridine (IdU) for 30 days, followed by a 40-day washout period. For labeling of cells that proliferate after injury, mice underwent knee surgery to produce an articular cartilage defect and received chlorodeoxyuridine (CIdU) for 4 days, starting at multiple time points after surgery. Unoperated and sham-operated joints served as controls. Knee joint paraffin sections were analyzed by double and triple immunostaining to detect nucleoside analogs, conventional MSC markers, and chondrocyte-lineage markers. RESULTS: Long-term-retaining, slow-cycling IdU-positive cells were detected in the synovium. At 4 days and 8 days after injury, there was marked proliferation of IdU-positive cells, which costained for CIdU. IdU-positive cells were nonhematopoietic, nonendothelial stromal cells, were distinct from pericytes, and stained positive for MSC markers. MSCs were phenotypically heterogeneous and located in topographically distinct niches in the lining layer and the subsynovial tissue. Twelve days after injury, double nucleoside-labeled cells within synovium were embedded in cartilage-specific metachromatic extracellular matrix and costained positive for the chondrocyte-lineage markers Sox9 and type II collagen. CONCLUSION: Our findings provide the first evidence of the existence of resident MSCs in the knee joint synovium that undergo proliferation and chondrogenic differentiation following injury in vivo.


Subject(s)
Chondrogenesis/physiology , Knee Joint/cytology , Mesenchymal Stem Cells/cytology , Stem Cell Niche/cytology , Synovial Membrane/cytology , Animals , Cell Count , Cell Proliferation , Immunohistochemistry , Knee Joint/physiology , Mice , Stem Cell Niche/physiology , Synovial Membrane/physiology
8.
Birth Defects Res C Embryo Today ; 90(4): 257-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21181887

ABSTRACT

Joint morphogenesis involves signaling pathways and growth factors that recur in the adult life with less redundancy to safeguard joint homeostasis. Loss of such homeostasis due to abnormal signaling networks as in aging could lead to diseases such as osteoarthritis. Stem cells are the cellular counterpart and targets of the morphogenetic signals, and they function to maintain the tissues by ensuring replacement of cells lost to physiological turnover, injury, aging, and disease. Mesenchymal stem cells (MSCs) are key players in regenerative medicine for their ability to differentiate toward multiple lineages such as cartilage and bone, but they age along the host body and senesce when serially passaged in culture. Understanding correlations between aging and its effects on MSCs is of the utmost importance to explain how aging happens and unravel the underlying mechanisms. The investigation of the MSC senescence in culture will help in developing more efficient and standardized cell culture methods for cellular therapies in skeletal regenerative medicine. An important area to explore in biomedical sciences is the role of endogenous stem cell niches in joint homeostasis, remodeling, and disease. It is anticipated that an understanding of the stem cell niches and related remodeling signals will allow the development of pharmacological interventions to support effective joint tissue regeneration, to restore joint homeostasis, and to prevent osteoarthritis.


Subject(s)
Aging/physiology , Homeostasis/physiology , Joints/growth & development , Mesenchymal Stem Cells/physiology , Osteoarthritis/pathology , Cartilage/physiology , Cell Differentiation , Humans , Joints/pathology , Joints/physiopathology , Musculoskeletal System/physiopathology , Regenerative Medicine , Stem Cells/cytology
9.
Diabetes ; 59(12): 3139-47, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20841611

ABSTRACT

OBJECTIVE: A number of clinical trials are underway to test whether mesenchymal stem cells (MSCs) are effective in treating various diseases, including type 1 diabetes. Although this cell therapy holds great promise, the optimal source of MSCs has yet to be determined with respect to major histocompatibility complex matching. Here, we examine this question by testing the ability of congenic MSCs, obtained from the NOR mouse strain, to reverse recent-onset type 1 diabetes in NOD mice, as well as determine the immunomodulatory effects of NOR MSCs in vivo. RESEARCH DESIGN AND METHODS: NOR MSCs were evaluated with regard to their in vitro immunomodulatory function in the context of autoreactive T-cell proliferation and dendritic cell (DC) generation. The in vivo effect of NOR MSC therapy on reversal of recent-onset hyperglycemia and on immunogenic cell subsets in NOD mice was also examined. RESULTS: NOR MSCs were shown to suppress diabetogenic T-cell proliferation via PD-L1 and to suppress generation of myeloid/inflammatory DCs predominantly through an IL-6-dependent mechanism. NOR MSC treatment of experimental type 1 diabetes resulted in long-term reversal of hyperglycemia, and therapy was shown to alter diabetogenic cytokine profile, to diminish T-cell effector frequency in the pancreatic lymph nodes, to alter antigen-presenting cell frequencies, and to augment the frequency of the plasmacytoid subset of DCs. CONCLUSIONS: These studies demonstrate the inimitable benefit of congenic MSC therapy in reversing experimental type 1 diabetes. These data should benefit future clinical trials using MSCs as treatment for type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Mesenchymal Stem Cell Transplantation/methods , Animals , Antigens, CD/analysis , Antigens, CD/immunology , Autoantigens/immunology , Cell Differentiation , Cytokines/analysis , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred NOD , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/pathology
10.
Hum Gene Ther ; 21(10): 1226-38, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20804388

ABSTRACT

Mesenchymal stromal/stem cells (MSCs) are a population of stromal cells present in the bone marrow and most connective tissues, capable of differentiation into mesenchymal tissues such as bone and cartilage. MSCs are attractive candidates for biological cell-based tissue repair approaches because of their extensive proliferative ability in culture while retaining their mesenchymal multilineage differentiation potential. In addition to its undoubted scientific interest, the prospect of monitoring and controlling MSC differentiation is a crucial regulatory and clinical requirement. Hence, the molecular regulation of MSC differentiation has been extensively studied. Most of the studies are in vitro, because the identity of MSCs in their tissues of origin in vivo remains undefined. This review addresses the current knowledge of the molecular basis of differentiation of cultured MSCs, with a particular focus on chondrogenesis and osteogenesis. Building on the information coming from developmental biology studies of embryonic skeletogenesis, several signaling pathways and transcription factors have been investigated and shown to play critical roles in MSC differentiation. In particular, the Wnt and transforming growth factor-ß/bone morphogenetic protein signaling pathways are well known to modulate in MSCs the molecular differentiation into cartilage and bone. Relevant to the emerging concept of stem cell niches is the demonstration that physical factors can also participate in the regulation of MSC differentiation. Knowledge of the regulation of MSC differentiation will be critical in the design of three-dimensional culture systems and bioreactors for automated bioprocessing through mathematical models applied to systems biology and network science.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/cytology , Signal Transduction , Animals , Cell Differentiation/genetics , Cells, Cultured , Chondrogenesis , Humans , Mesenchymal Stem Cells/physiology , Mice , Osteogenesis , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
11.
J Immunol ; 184(6): 2939-48, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20164431

ABSTRACT

Ischemic (isc) injury during the course of transplantation enhances the immunogenicity of allografts and thus results in poorer graft outcome. Given the central role of dendritic cells (DCs) in mounting alloimmune responses, activation of donor DCs by ischemia may have a primary function in the increased immunogenicity of isc allografts. In this study, we sought to investigate the effect of ischemia on DC activity in vitro. Following induction of ischemia, bone marrow-derived DCs were shown to augment allogeneic T cell proliferation as well as the IFN-gamma response. Isc DCs produced greater levels of IL-6, and isc insult was concurrent with NF-kappaB activation. TLR4 ligation was also shown to occur in isc DCs, most likely in response to the endogenous ligand heat shock protein 70, which was found to be elevated in DCs following isc injury, and lack of TLR4 abrogated the observed effects of isc DCs. As compared with control DCs, isc DCs injected into the footpads of mice demonstrated enhanced migration, which was concomitant with increased recipient T cell activity. Moreover, isc DCs underwent a greater degree of apoptosis in the lymph nodes of injected mice, which may further demonstrate enhanced immunogenicity of isc DCs. We thus show that isc injury of DCs enhances DC function, augments the allogeneic T cell response, and occurs via ligation of TLR4, followed by activation of NF-kappaB. These data may serve to identify novel therapeutic targets to attenuate graft immunogenicity following ischemia.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/pathology , Ischemia/chemically induced , Ischemia/immunology , Mineral Oil/toxicity , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Up-Regulation/immunology , Animals , Cells, Cultured , Coculture Techniques , Dendritic Cells/drug effects , Injections, Intraperitoneal , Ischemia/pathology , Ligands , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/deficiency , Up-Regulation/genetics
12.
Eur Cell Mater ; 20: 121-33, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-21249629

ABSTRACT

Mesenchymal Stromal Progenitor/Stem Cells (MSCs) are a rare population of non-hematopoietic stromal cells, present in the bone marrow and most connective tissues of the body. They are capable of differentiation into mesenchymal tissues such as bone, cartilage, adipose tissue and muscle. In the absence of specific markers, MSCs have been defined following isolation and culture expansion, by their expression of various molecules including CD90, CD105 and CD73 and absence of markers like CD34, CD45, and CD14. MSCs have extensive proliferative ability in culture in an uncommitted state while retaining their multilineage differentiation potential, which make them attractive candidates for biological cell-based tissue repair approaches. However, their identity in their tissues of origin is not clear and the niches in which they reside are not defined. This review addresses the current state of MSC research including the differentiation potency of culture expanded MSCs, expression of chemokines and their receptors in MSCs--both relevant issues for the advocated use of MSCs for tissue repair and their systemic delivery to the affected tissues. It also reviews current knowledge of MSC niches in their native tissues, addressing the relationship with pericytes. Finally, it provides a scientific basis for the requirement of a thorough characterisation of the endogenous MSC niches within their native tissues in vivo. The knowledge of MSC niches will instruct development of innovative therapeutic measures such as producing pharmacological substances that target endogenous MSCs and their niches in order to activate and guide intrinsic repair and to improve disease outcomes.


Subject(s)
Cell Movement , Mesenchymal Stem Cells/physiology , Stem Cell Niche/physiology , Animals , Cell Differentiation , Cells, Cultured , Chemokines/biosynthesis , Chondrogenesis , Guided Tissue Regeneration , Humans , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Osteogenesis , Receptors, Chemokine/biosynthesis , Stem Cell Niche/cytology
13.
J Immunol ; 183(2): 993-1004, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19561093

ABSTRACT

Human clinical trials in type 1 diabetes (T1D) patients using mesenchymal stem cells (MSC) are presently underway without prior validation in a mouse model for the disease. In response to this void, we characterized bone marrow-derived murine MSC for their ability to modulate immune responses in the context of T1D, as represented in NOD mice. In comparison to NOD mice, BALB/c-MSC mice were found to express higher levels of the negative costimulatory molecule PD-L1 and to promote a shift toward Th2-like responses in treated NOD mice. In addition, transfer of MSC from resistant strains (i.e., nonobese resistant mice or BALB/c), but not from NOD mice, delayed the onset of diabetes when administered to prediabetic NOD mice. The number of BALB/c-MSC trafficking to the pancreatic lymph nodes of NOD mice was higher than in NOD mice provided autologous NOD-MSC. Administration of BALB/c-MSC temporarily resulted in reversal of hyperglycemia in 90% of NOD mice (p = 0.002). Transfer of autologous NOD-MSC imparted no such therapeutic benefit. We also noted soft tissue and visceral tumors in NOD-MSC-treated mice, which were uniquely observed in this setting (i.e., no tumors were present with BALB/c- or nonobese resistant mice-MSC transfer). The importance of this observation remains to be explored in humans, as inbred mice such as NOD may be more susceptible to tumor formation. These data provide important preclinical data supporting the basis for further development of allogeneic MSC-based therapies for T1D and, potentially, for other autoimmune disorders.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Animals , Bone Marrow Cells , Cell Movement , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/immunology , Hyperglycemia/therapy , Immunologic Factors/immunology , Mesenchymal Stem Cell Transplantation/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Neoplasms/etiology , Treatment Outcome
14.
Tissue Eng Part A ; 15(8): 2203-12, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19265473

ABSTRACT

The contribution of the host's circulating progenitor cells after implantation of mesenchymal stem cells (MSC)/bioscaffold combinations for repairing bone defects has not been elucidated, although this issue affects the clinical application of the tissue engineering approach. We implanted blocks of hydroxyapatite loaded with murine MSCs into syngenic, allogenic, and immunocompromised recipients. After 8 weeks, we found that bone tissue was formed in syngenic and immunocompromised animals. The implanted cells appeared pivotal in the early stages of tissue development, but cells of the recipient's origin finally made bone. In this system, osteoprogenitors migrated from the recipient to the implant, whereas the implanted cells left the scaffold and entered the circulatory flow. We observed rapid destruction of implanted cells when allogenic MSC/bioscaffold combinations were grafted onto immunocompetent recipients without immunosuppressant therapy. This destruction blocked the recruitment process and did not allow the formation of new bone tissue. The possibility that the implanted exogenous MSCs could engage the host's osteoprogenitor cells to form new bone tissue could open new perspectives for the tissue engineering approach to bone repair, including the opportunity of using allogenic cells combined with a temporary immunosuppressant therapy, stimulating the replacement of the exogenous cells with autologous cells.


Subject(s)
Ceramics/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Animals , Graft Rejection , Immunocompetence , Immunophenotyping , Implants, Experimental , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Porosity/drug effects , Tissue Scaffolds , Transplantation, Homologous
15.
J Am Soc Nephrol ; 20(3): 535-44, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129312

ABSTRACT

Little is known about heart tissue/donor dendritic cells, which play a key role in mounting alloimmune responses. In this report, we focus on three primary features of donor dendritic cells: their generation, their trafficking after transplantation, and their role in regulating tolerance versus rejection. Using transgenic mice as donors of heart allografts enabled us to monitor trafficking of donor dendritic cells after transplantation. Donor dendritic cells rapidly migrated into secondary lymphoid tissues within 3 h of transplantation. We found that the chemokine receptor CX3CR1 regulates the generation of heart tissue dendritic cells constitutively. Compared with wild-type hearts, CX3CR1(-/-) hearts contained fewer dendritic cells, and heart allografts from CX3CR1(-/-) donors survived significantly longer without immunosuppression. Unexpectedly, though, co-stimulatory blockade with anti-CD154 or CTLA4-Ig induced long-term survival for wild-type heart allografts but not for CX3CR1(-/-) heart allografts. Increasing the dendritic cell frequency in CX3CR1(-/-) hearts by treatment with Flt3L restored the anti-CD154-induced prolongation of CX3CR1(-/-) heart allograft survival. Compared with wild-type donors, depleting transgenic donors of dendritic cells before heart transplantation also markedly worsened chronic rejection under anti-CD154 treatment. These data indicate the importance of the CX3CR1 pathway in the generation of heart tissue dendritic cells and the divergent role of tissue/dendritic cells in rejection versus tolerance.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Graft Rejection/immunology , Heart Transplantation/immunology , Transplantation Tolerance/immunology , Animals , CD40 Ligand/antagonists & inhibitors , Green Fluorescent Proteins/genetics , Isoantibodies/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-8A/deficiency , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/physiology , Recombinant Proteins/genetics , T-Lymphocytes/immunology , Tissue Donors , Transplantation, Homologous
16.
Carcinogenesis ; 30(1): 150-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18849298

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSCs) are precursors of bone, cartilage and fat tissue. MSC can also regulate the immune response. For these properties, they are tested in clinical trials for tissue repair in combination with bioscaffolds or injected as cell suspension for immunosuppressant therapy. Experimental data, however, indicate that MSC can undergo or induce a tumorigenic process in determined circumstances. We used a modified model of ectopic bone formation in mice by subcutaneously implanting porous ceramic seeded with murine MSC. In this new model, host-derived sarcomas developed when we implanted MSC/bioscaffold constructs into syngeneic and immunodeficient recipients, but not in allogeneic hosts or when MSCs were injected as cell suspensions. The bioscaffold provided a tridimensional support for MSC to aggregate, thus producing the stimulus for triggering the process eventually leading to the transformation of surrounding cells and creating a surrogate tumor stroma. The chemical and physical characteristics of the bioscaffold did not affect tumor formation; sarcomas developed either when a stiff porous ceramic was used or when the scaffold was a smooth collagen sponge. The immunoregulatory function of MSC contributed to tumor development. Implanted MSC expanded clones of CD4+CD25+ T regulatory lymphocytes that suppressed host's antitumor immune response.


Subject(s)
Cell Transplantation , Mesenchymal Stem Cells/cytology , Sarcoma, Experimental/pathology , Animals , Cell Proliferation , Green Fluorescent Proteins/genetics , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Sarcoma, Experimental/immunology
17.
Transplantation ; 85(7): 1030-8, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18408585

ABSTRACT

BACKGROUND: Trafficking of dendritic cells (DC), the primary regulators of alloimmune responses, is controlled by chemokines. Here, we provide evidence that lack of CCR2 could lead to the generation of functionally and phenotypically different DC, which in part could explain the benefits observed in transplanting islets in CCR2 recipients. METHODS AND RESULTS: We show that, in contrast to the in vitro DC maturation model, in vivo DC maturation is accompanied by an increase in the expression of CCR2. Compared with wild-type (WT), DC generated in vitro from CCR2 mice, and DC extracted from CCR2 naïve mice or from CCR2 recipients of islet allografts, display lesser allostimulatory capacity. Compared with WT DC, CCR2 DC produce more IL-4 and induce more IL-4-producing T cells. CCR2 DC also promote the generation of regulatory T cells that more efficiently suppress T cell proliferative responses by mixed leukocyte reaction. Similarly, the percentage of CD4CD25FoxP3 cells were found to be higher in CCR2 recipients of islet allografts than in WT recipients. CONCLUSIONS: In summary, lack of CCR2 interferes with the allostimulatory capacity of DC and promotes the generation of regulatory T cells. This is the first demonstration of a mechanistic link between targeting a specific chemokine pathway and the DC-regulatory T cell axis in alloimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/surgery , Graft Rejection/immunology , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Cell Movement , Diabetes Mellitus, Experimental/immunology , Flow Cytometry , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Phenotype , Transplantation, Homologous
18.
Invest Ophthalmol Vis Sci ; 49(5): 1957-70, 2008 May.
Article in English | MEDLINE | ID: mdl-18281616

ABSTRACT

PURPOSE: Experimental autoimmune uveitis (EAU) is an established model for immune-mediated human uveitis. Although several genes from major histocompatibility complex (MHC) loci have been shown to play a role in uveitis, little is known about the role of non-MHC genes in the pathogenesis of EAU. Several non-MHC genes have been implicated in the pathogenesis of various autoimmune diseases. The primary objective of this study was to identify the non-MHC genes involved in the pathogenesis of EAU, to identify potential drug targets and possibly to target their protein products for immunotherapy. METHODS: EAU was induced in the susceptible (Lewis; LEW) or resistant (Fischer 344; F344) rats that have identical MHC class II haplotype. Draining lymph node cells were obtained during the innate and adaptive phase of the immune response, and the pattern of gene expression was evaluated using microarray technology. Differentially expressed genes were validated at mRNA and protein levels using various methods. RESULTS: Susceptibility to EAU was associated with an increased expression of numerous non-MHC genes such as Th1-type cytokines and chemokines, antiapoptotic factors, hormones, and neurotransmitters and a downregulation of selected adhesion molecules. In this study a combined genetic-genomic approach was used to identify different patterns of gene expression associated with the sensitization and effector phase of EAU pathogenesis. CONCLUSIONS: The data demonstrate that the differential expression of several non-MHC genes is associated with the mechanism of uveitis.


Subject(s)
Autoimmune Diseases/genetics , Disease Models, Animal , Gene Expression Regulation/physiology , Genes, MHC Class II/physiology , Uveitis/genetics , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Autoimmune Diseases/immunology , Disease Susceptibility , Eye Proteins/genetics , Female , Flow Cytometry , Gene Expression Profiling , Lymphocyte Activation , Oligonucleotide Array Sequence Analysis , Peptide Fragments , Quantitative Trait Loci , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Retinol-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Uveitis/immunology
19.
Arthritis Rheum ; 56(4): 1175-86, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17393437

ABSTRACT

OBJECTIVE: Mesenchymal stem cells (MSCs) are precursors of tissue of mesenchymal origin, but they also have the capacity to regulate the immune response by suppressing T and B lymphocyte proliferation in a non-major histocompatibility complex-restricted manner. Use of MSCs as immunosuppressant agents in autoimmune diseases has been proposed and successfully tested in animal models. We explored the feasibility of using allogeneic MSCs as therapy for collagen-induced arthritis, a mouse model for human rheumatoid arthritis. METHODS: DBA/1 mice were immunized with type II collagen in Freund's complete adjuvant, and some of the animals received an intraperitoneal injection of allogeneic MSCs. RESULTS: A single injection of MSCs prevented the occurrence of severe, irreversible damage to bone and cartilage. MSCs induced hyporesponsiveness of T lymphocytes as evidenced by a reduction in active proliferation, and modulated the expression of inflammatory cytokines. In particular, the serum concentration of tumor necrosis factor alpha was significantly decreased. MSCs exerted their immunomodulatory function by educating antigen-specific Tregs. CONCLUSION: Our results suggest an effective new therapeutic approach to target the pathogenic mechanism of autoimmune arthritis using allogeneic MSCs. However, further studies are required before these results can be translated to clinical settings.


Subject(s)
Arthritis, Experimental/therapy , Bone and Bones/drug effects , Cartilage, Articular/drug effects , Joints/drug effects , Mesenchymal Stem Cell Transplantation , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Bone Marrow Cells , Bone and Bones/pathology , Cartilage, Articular/pathology , Cell Proliferation/drug effects , Cytokines/blood , Female , Joints/pathology , Male , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transplantation, Homologous
20.
Diabetes ; 56(4): 912-20, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17287465

ABSTRACT

Dendritic cells (DCs) are the most potent antigen-presenting cells, yet little data are available on the differential characteristics of donor and recipient DCs (dDCs and rDCs, respectively) during the process of islet allograft rejection. DTR-GFP-DC mice provide a novel tool to monitor DC trafficking and characteristics during allograft rejection. We show rapid migration of dDCs to recipient lymphoid tissues as early as 3 h post-islet allotransplantation. Compared with rDCs, dDCs express different patterns of chemokine receptors, display differential proliferative capacity, and exhibit a higher level of maturity; these findings could be attributed to the effects of injury that dDCs undergo during islet cell preparation and engraftment. Intriguingly, we detected dDCs in the spleen of recipients long after rejection of islet allografts. Given that dDCs express high levels of CCR7, islets were cultured before transplant with the ligand for CCR7 (CCL21). This novel method, which enabled us to enhance the efflux of dDCs from islet preparations, resulted in a prolongation of islet allograft survival in immunocompetent recipients. This study introduces dDCs and rDCs as two distinct types of DCs and provides novel data with clinical implications to use chemokine-based DC-depleting strategies to prolong islet allograft survival.


Subject(s)
Chemokines, CC/physiology , Dendritic Cells/cytology , Dendritic Cells/transplantation , Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/physiology , Animals , Bone Marrow Cells/cytology , Cell Division , Chemokine CCL21 , Dendritic Cells/drug effects , Genes, Reporter , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...