Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Int J Mol Sci ; 22(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34681764

ABSTRACT

Vascularized composite allografts contain various tissue components and possess relative antigenicity, eliciting different degrees of alloimmune responses. To investigate the strategies for achieving facial allograft tolerance, we established a mouse hemiface transplant model, including the skin, muscle, mandible, mucosa, and vessels. However, the immunomodulatory effects of the mandible on facial allografts remain unclear. To understand the effects of the mandible on facial allograft survival, we compared the diversities of different facial allograft-elicited alloimmunity between a facial osteomyocutaneous allograft (OMC), including skin, muscle, oral mucosa, and vessels, and especially the mandible, and a myocutaneous allograft (MC) including the skin, muscle, oral mucosa, and vessels, but not the mandible. The different facial allografts of a BALB/c donor were transplanted into a heterotopic neck defect on fully major histocompatibility complex-mismatched C57BL/6 mice. The allogeneic OMC (Allo-OMC) group exhibited significant prolongation of facial allograft survival compared to the allogeneic MC group, both in the presence and absence of FK506 immunosuppressive drugs. With the use of FK506 monotherapy (2 mg/kg) for 21 days, the allo-OMC group, including the mandible, showed prolongation of facial allograft survival of up to 65 days, whereas the myocutaneous allograft, without the mandible, only survived for 34 days. The Allo-OMC group also displayed decreased lymphocyte infiltration into the facial allograft. Both groups showed similar percentages of B cells, T cells, natural killer cells, macrophages, and dendritic cells in the blood, spleen, and lymph nodes. However, a decrease in pro-inflammatory T helper 1 cells and an increase in anti-inflammatory regulatory T cells were observed in the blood and lymph nodes of the Allo-OMC group. Significantly increased percentages of donor immune cells were also observed in three lymphoid organs of the Allo-OMC group, suggesting mixed chimerism induction. These results indicated that the mandible has the potential to induce anti-inflammatory effects and mixed chimerism for prolonging facial allograft survival. The immunomodulatory understanding of the mandible could contribute to reducing the use of immunosuppressive regimens in clinical face allotransplantation including the mandible.


Subject(s)
Facial Transplantation/adverse effects , Graft Rejection/etiology , Mandible/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Facial Transplantation/methods , Graft Rejection/immunology , Graft Survival/physiology , Immunosuppressive Agents/pharmacology , Mandible/immunology , Mandible/transplantation , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin Transplantation/adverse effects , Skin Transplantation/methods , Tacrolimus/pharmacology , Transplantation Chimera/physiology , Transplantation, Homologous
2.
Front Immunol ; 11: 1028, 2020.
Article in English | MEDLINE | ID: mdl-32536926

ABSTRACT

B cell adaptor molecule of 32 kDa (Bam32), known as dual adapter for phosphotyrosine and 3-phosphoinositides 1 (DAPP1), has been implicated in regulating lymphocyte proliferation and recruitment during inflammation. However, its role in neutrophils during inflammation remains unknown. Using intravital microscopy, we examined the role of Bam32 in formyl peptide receptor agonist WKYMVm-induced permeability changes in post-capillary venules and assessed simultaneously neutrophil adhesion and emigration in cremaster muscles of Bam32-deficient (Bam32-/-) and wild-type (WT) control mice. We observed significantly reduced WKYMVm-induced microvascular hyperpermeability accompanied by markedly decreased neutrophil emigration in Bam32-/- mice. The Bam32-specific decrease in WKYMVm-induced hyperpermeability was neutrophil-dependent as this was verified in bone marrow transplanted chimeric mice. We discovered that Bam32 was critically required for WKYMVm-induced intracellular and extracellular production of reactive oxygen species (ROS) in neutrophils. Pharmacological scavenging of ROS eliminated the differences in WKYMVm-induced hyperpermeability between Bam32-/- and WT mice. Deficiency of Bam32 decreased WKYMVm-induced ERK1/2 but not p38 or JNK phosphorylation in neutrophils. Inhibition of ERK1/2 signaling cascade suppressed WKYMVm-induced ROS generation in WT neutrophils and microvascular hyperpermeability in WT mice. In conclusion, our study reveals that Bam32-dependent, ERK1/2-involving ROS generation in neutrophils is critical in WKYMVm-induced microvascular hyperpermeability during neutrophil recruitment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Lipoproteins/metabolism , Neutrophils/metabolism , Oligopeptides/pharmacology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Transplantation , Capillary Permeability/immunology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion/physiology , Lipoproteins/deficiency , Lipoproteins/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Neutrophil Infiltration/physiology , Neutrophils/drug effects , Neutrophils/immunology , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/agonists , Transplantation Chimera/immunology , Transplantation Chimera/physiology , Venules/drug effects , Venules/immunology , Venules/physiology
3.
Sci Rep ; 9(1): 19434, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31857640

ABSTRACT

The mouse (Mus musculus) is an extensively used model of human disease and responses to stresses such as ionizing radiation. As part of our work developing gene expression biomarkers of radiation exposure, dose, and injury, we have found many genes are either up-regulated (e.g. CDKN1A, MDM2, BBC3, and CCNG1) or down-regulated (e.g. TCF4 and MYC) in both species after irradiation at ~4 and 8 Gy. However, we have also found genes that are consistently up-regulated in humans and down-regulated in mice (e.g. DDB2, PCNA, GADD45A, SESN1, RRM2B, KCNN4, IFI30, and PTPRO). Here we test a hematopoietically humanized mouse as a potential in vivo model for biodosimetry studies, measuring the response of these 14 genes one day after irradiation at 2 and 4 Gy, and comparing it with that of human blood irradiated ex vivo, and blood from whole body irradiated mice. We found that human blood cells in the hematopoietically humanized mouse in vivo environment recapitulated the gene expression pattern expected from human cells, not the pattern seen from in vivo irradiated normal mice. The results of this study support the use of hematopoietically humanized mice as an in vivo model for screening of radiation response genes relevant to humans.


Subject(s)
Gene Expression Regulation/radiation effects , Hematopoietic Stem Cell Transplantation , Models, Animal , Transplantation Chimera/physiology , Whole-Body Irradiation/adverse effects , Animals , Biomarkers/analysis , Dose-Response Relationship, Radiation , Down-Regulation/physiology , Down-Regulation/radiation effects , Humans , Male , Mice , Species Specificity , Up-Regulation/physiology , Up-Regulation/radiation effects
4.
Stem Cells Dev ; 27(23): 1599-1604, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30319057

ABSTRACT

The search for a better animal model to simulate human disease has been a "holy grail" of biomedical research for decades. Recent identification of different types of pluripotent stem (PS) cells and advances in chimera research might soon permit the generation of interspecies chimeras from closely related species, such as those between humans and other primates. In this study, we suggest that the creation of human-primate chimeras-specifically, the transfer of human stem cells into (non-ape) primate hosts-could not only surpass the limitations of current monkey models of neurological and psychiatric disease but would also raise important ethical considerations concerning the use of monkeys in invasive research. Questions regarding the scientific value and ethical concerns raised by the prospect of human-monkey chimeras are more urgent in light of recent advances in PS cell research and attempts to generate interspecies chimeras between humans and animals. While some jurisdictions prohibit the introduction of human PS cells into monkey preimplantation embryos, other jurisdictions may permit and even encourage such experiments. Therefore, it is useful to consider blastocyst complementation experiments more closely in light of advances that could make these chimeras possible and to consider the ethical and political issues that are raised.


Subject(s)
Disease Models, Animal , Haplorhini/genetics , Mental Disorders/genetics , Neurodegenerative Diseases/genetics , Transplantation Chimera/genetics , Animals , Embryo Research/ethics , Haplorhini/physiology , Humans , Mental Disorders/pathology , Neurodegenerative Diseases/pathology , Stem Cell Transplantation/ethics , Stem Cell Transplantation/methods , Stem Cell Transplantation/standards , Transplantation Chimera/physiology
5.
J Heart Lung Transplant ; 37(9): 1119-1130, 2018 09.
Article in English | MEDLINE | ID: mdl-29699851

ABSTRACT

BACKGROUND: The mixed chimerism approach for intentional induction of donor-specific tolerance was shown to be successful in various models from mice to humans. For transplant patients, the approach would obviate the need for long-term immunosuppression and associated side effects; moreover, it would preclude the risk of late graft loss due to chronic rejection. Widespread clinical application is hindered by toxicities related to recipient pre-conditioning. Herein we aimed to investigate a clinically relevant protocol for tolerance induction to cardiac allografts, sparing CD40 blockade or T-cell depletion. METHODS: B6 mice were conditioned with non-myeloablative total body irradiation, fully mismatched BALB/c bone marrow cells, and short-term therapy, based on either anti- lymphocyte function-associated antigen-1 (anti-LFA-1) or anti-CD40L. Multilineage chimerism was followed by flow-cytometric analysis, tolerance was assessed with skin and heart allografts from fully or major histocompatibility complex-mismatched donors. Mechanisms of tolerance were investigated by analysis of donor-specific antibodies (DSAs), mixed lymphocyte reaction (MLR) assays, and deletion of donor-reactive T cells. RESULTS: We found that the combination of cytotoxic T-lymphocyte antigen 4 immunoglobulin (CTLA4Ig) and rapamycin with LFA-1 blockade enhanced bone marrow engraftment and led to more efficient T-cell engraftment and subsequent tolerization. Although fully mismatched skin grafts were chronically rejected, primarily vascularized heart allografts survived indefinitely and without signs of chronic rejection, independent of minor antigen mismatches. CONCLUSIONS: We have demonstarted a robust protocol for the induction of tolerance for cardiac allografts in the absence of CD40 blockade. Our findings demonstrate the potential of a clinically relevant minimal conditioning protocol designed to induce lifelong immunologic tolerance toward cardiac allografts.


Subject(s)
Graft Rejection/physiopathology , Graft Survival/physiology , Lymphocyte Function-Associated Antigen-1/physiology , T-Lymphocytes/physiology , Transplantation Chimera/physiology , Animals , CD40 Antigens/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred Strains
6.
J Vis Exp ; (127)2017 09 06.
Article in English | MEDLINE | ID: mdl-28930976

ABSTRACT

The presence of incomplete chimerism is noted in a large proportion of patients following bone marrow transplant for thalassemia major or sickle cell disease. This observation has tremendous implications, as subsequent therapeutic immunomodulation strategies can improve clinical outcome. Conventionally, polymerase chain reaction-based analysis of short tandem repeats is used to identify chimerism in donor-derived blood cells. However, this method is restricted to nucleated cells and cannot distinguish between dissociated single-cell lineages. We applied the analysis of short tandem repeats to flow cytometric-sorted hematopoietic progenitor cells and compared this with the analysis of short tandem repeats obtained from selected burst-forming unit - erythroid colonies, both collected from the bone marrow. With this method we are able to demonstrate the different proliferation and differentiation of donor cells in the erythroid compartment. This technique is eligible to complete current monitoring of chimerism in the stem cell transplant setting and thus may be applied in future clinical studies, stem cell research and design of gene therapy trials.


Subject(s)
Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/transplantation , Hematopoietic Stem Cell Transplantation/methods , Hemoglobinopathies/therapy , Transplantation Chimera/physiology , Hemoglobinopathies/pathology , Humans , Transplantation Conditioning/methods
7.
Methods Mol Biol ; 1506: 91-100, 2017.
Article in English | MEDLINE | ID: mdl-27830547

ABSTRACT

Primary or cryopreserved human hepatocytes (h-heps) have been used as the gold standard for in vitro metabolism and hepatotoxicity studies; however, the supply of h-heps is limited and they cannot grow in vitro. We achieved approximately 1000-fold propagation of h-heps in the liver of albumin promoter/enhancer-driven urokinase-type plasminogen activator transgenic/severe combined immunodeficiency disease (uPA/SCID) mice with genetically induced liver disease and immunodeficiency. When h-heps are transplanted into the uPA/SCID mouse liver via the spleen, the h-heps engraft in the mouse liver, resulting in its repopulation with h-heps. We have named this model "chimeric mouse with humanized liver, PXB-mouse®." Fresh h-heps can be isolated from the chimeric mice (PXB-cells®) and have been used for in vitro studies.The efficacy and safety of chemical entities for use in humans are estimated using experimental animals such as rats and mice. The drug development of many chemical entities has been halted because of metabolic differences between humans and animals during clinical studies. Therefore, chimeric mice with humanized liver have been used to predict human-type metabolism and safety conditions for h-heps. In addition, until recently there were no suitable hepatitis B or C virus (HBV or HCV) susceptible animal models aside from chimpanzees. Chimeric mice are the sole persistent infectious small animal model for HBV and HCV and they have been used to investigate the efficacy of new anti-HBV or HCV agents.In this chapter, we describe a method for producing chimeric mice with humanized liver using uPA/SCID mice.


Subject(s)
Cell Transplantation/methods , Disease Models, Animal , Hepatocytes/transplantation , Liver Diseases/genetics , Transplantation Chimera/physiology , Albumins/genetics , Animals , Female , Hepatocytes/physiology , Humans , Liver/cytology , Liver/surgery , Liver Diseases/surgery , Male , Mice , Mice, SCID , Mice, Transgenic , Promoter Regions, Genetic/genetics , Transplantation, Heterologous , Urokinase-Type Plasminogen Activator/genetics
8.
Methods Mol Biol ; 1506: 117-130, 2017.
Article in English | MEDLINE | ID: mdl-27830549

ABSTRACT

Mice with humanized chimeric liver are promising in vivo tools to evaluate the efficacy of novel compounds or vaccine induced antibodies directed against pathogens that infect the human liver. In addition they can be used to study the human-type metabolism of medicinal compounds and hepatotoxicity.


Subject(s)
Cell Transplantation/methods , Chemical and Drug Induced Liver Injury/surgery , Hepatocytes/transplantation , Transplantation Chimera/physiology , Acetoacetates/metabolism , Acetoacetates/toxicity , Animals , Chemical and Drug Induced Liver Injury/genetics , Disease Models, Animal , Humans , Hydrolases/genetics , Liver/cytology , Liver/metabolism , Liver/surgery , Mice , Mice, Knockout , Mice, SCID , Transplantation Chimera/genetics , Urokinase-Type Plasminogen Activator/genetics
9.
Methods Mol Biol ; 1506: 179-192, 2017.
Article in English | MEDLINE | ID: mdl-27830553

ABSTRACT

Hepatocyte transplantation is the best approach to maintain and propagate differentiated hepatocytes from different species. Host liver has to be adapted for transplanted hepatocytes productive engraftment and proliferation being required a chronic liver injury to eliminate host hepatocytes and provide a proliferative advantage to the transplanted hepatocytes. Most valuable mouse models for xenograft hepatocyte transplantation are based on genetically modified animals to cause a chronic liver damage and to limit host hepatocyte regeneration potential. We present a methodology that generates a chronic liver damage and can be applied to any host mouse strain and animal species based on the inoculation of a recombinant adenovirus to express herpes simplex thymidine kinase in host hepatocytes sensitizing them to ganciclovir treatment. This causes a prolonged liver damage that allows hepatocyte transplantation and generation of regenerative nodules in recipient mouse liver integrated by transplanted cells and host sinusoidal. Obtained chimeric animals maintain functional chimeric nodules for several weeks, ready to be used in any study.


Subject(s)
Adenoviridae/genetics , Cell Transplantation/methods , Hepatocytes/transplantation , Liver Regeneration/drug effects , Liver/physiology , Transplantation Conditioning/methods , Animals , Cell Separation/methods , Cell Transplantation/adverse effects , Cell Transplantation/instrumentation , Chemical and Drug Induced Liver Injury, Chronic , Disease Models, Animal , Ganciclovir/toxicity , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Simplexvirus/genetics , Thymidine Kinase/genetics , Transduction, Genetic/methods , Transplantation Chimera/physiology , Transplantation Chimera/surgery , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/methods , Viral Nonstructural Proteins/genetics
10.
Stem Cells ; 33(11): 3228-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26138940

ABSTRACT

To date no authentic embryonic stem cell (ESC) line or germline-competent-induced pluripotent stem cell (iPSC) line has been established for large animals. Despite this fact, there is an impression in the field that large animal ESCs or iPSCs are as good as mouse counterparts. Clarification of this issue is important for a healthy advancement of the stem cell field. Elucidation of the causes of this failure in obtaining high quality iPSCs/ESCs may offer essential clues for eventual establishment of authentic ESCs for large animals including humans. To this end, we first generated porcine iPSCs using nonintegrating replicating episomal plasmids. Although these porcine iPSCs met most pluripotency criteria, they could neither generate cloned piglets through nuclear transfer, nor contribute to later stage chimeras through morula injections or aggregations. We found that the reprogramming genes in iPSCs could not be removed even under negative selection, indicating they are required to maintain self-renewal. The persistent expression of these genes in porcine iPSCs in turn caused differentiation defects in vivo. Therefore, incomplete reprogramming manifested by a reliance on sustained expression of exogenous-reprogramming factors appears to be the main reason for the inability of porcine iPSCs to form iPSC-derived piglets.


Subject(s)
Genetic Vectors/physiology , Induced Pluripotent Stem Cells/physiology , Plasmids/physiology , Transgenes/physiology , Transplantation Chimera/physiology , Animals , Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Female , Induced Pluripotent Stem Cells/transplantation , Mice , Mice, Nude , Nuclear Transfer Techniques , Swine , Swine, Miniature
11.
FASEB J ; 29(1): 105-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25342128

ABSTRACT

Heme oxygenase (HO)-2 deficiency impairs wound healing and exacerbates inflammation following injury. We examine the impact of HO-2 deficiency on macrophage function and the contribution of macrophage HO-2 to inflammatory and repair responses to injury. Corneal epithelial debridement was performed in control and macrophage-depleted HO-2(-/-) and wild-type (WT) mice and in bone marrow chimeras. Peritoneal macrophages were collected for determination of phagocytic activity and classically activated macrophage (M1)-alternatively activated macrophage (M2) polarization. Depletion of macrophages delayed corneal healing (13.2%) and increased neutrophil infiltration (54.1%) by day 4 in WT mice, whereas in HO-2(-/-) mice, it did not worsen the already impaired wound healing and exacerbated inflammation. HO-2(-/-) macrophages displayed an altered M1 phenotype with no significant expression of M2 or M2-like activated cells and a 31.3% reduction in phagocytic capacity that was restored by inducing HO-1 activity or supplementing biliverdin. Macrophage depletion had no effect, whereas adoptive transfer of WT bone marrow improved wound healing (34% on day 4) but did not resolve the exaggerated inflammatory response in HO-2(-/-) mice. These findings indicate that HO-2-deficient macrophages are dysfunctional and that macrophage HO-2 is required for proper macrophage function but is insufficient to correct the impaired healing of the HO-2(-/-) cornea, suggesting that corneal epithelial expression of HO-2 is a key to resolution and repair in wound healing.


Subject(s)
Corneal Injuries/physiopathology , Heme Oxygenase (Decyclizing)/deficiency , Macrophages/enzymology , Macrophages/physiology , Wound Healing/physiology , Animals , Bone Marrow Transplantation , Corneal Injuries/pathology , Cytokines/biosynthesis , Epithelium, Corneal/pathology , Epithelium, Corneal/physiopathology , Female , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/pathology , Inflammation/physiopathology , Macrophage Activation , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/physiology , Phagocytosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transplantation Chimera/physiology
12.
Microsurgery ; 35(1): 45-51, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25073635

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) induces angiogenesis and osteogenesis in bone allotransplants. We aim to determine whether bone remodeling in VEGF-treated bone allotransplants results from repopulation with circulation-derived autogenous cells or survival of allogenic transplant-derived cells. METHODS: Vascularized femoral bone transplants were transplanted from female Dark Agouti rats (DA;RT1(a) ) to male Piebald Viral Glaxo (PVG;RT1(c) ). Arteriovenous bundle implantation and short-term immunosuppression were used to maintain cellular viability. VEGF was encapsulated in biodegradable microspheres and delivered intramedullary in the experimental group (n = 22). In the control group (n = 22), no VEGF was delivered. Rats were sacrificed at 4 or 18 weeks. Laser capture microdissection of bone remodeling areas was performed at the inner and outer cortex. Sex-mismatched genes were quantified with reverse transcription-polymerase chain reaction to determine the amount of male cells to total cells, defined as the relative expression ratio (rER). RESULTS: At 4 weeks, rER was significantly higher at the inner cortex in VEGF-treated transplants as compared to untreated transplants (0.622 ± 0.225 vs. 0.362 ± 0.081, P = 0.043). At 4 weeks, the outer cortex in the control group had a significantly higher rER (P = 0.038), whereas in the VEGF group, the inner cortex had a higher rER (P = 0.015). Over time, in the outer cortex the rER significantly increased to 0.634 ± 0.106 at 18 weeks in VEGF-treated rats (P = 0.049). At 18 weeks, the rER was >0.5 at all cortical areas in both groups. CONCLUSIONS: These in vivo findings suggest a chemotactic effect of intramedullary applied VEGF on recipient-derived bone and could imply that more rapid angiogenesis of vascularized allotransplants can be established with microencapsulated VEGF.


Subject(s)
Bone Transplantation/methods , Bone and Bones/blood supply , Microsurgery , Transplantation Chimera/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Capsules , Female , Male , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Rats , Rats, Inbred Strains , Vascular Endothelial Growth Factor A/administration & dosage
13.
Chin J Dent Res ; 17(2): 105-10, 2014.
Article in English | MEDLINE | ID: mdl-25531018

ABSTRACT

OBJECTS: To test the hypothesis whether bone marrow stem cells (BMSCs) could migrate into the periodontium as the precursor available for the repair of tissue injury. METHODS: A chimeric mouse model was established by transplanting BMSCs derived from red fluorescent protein mouse into irradiated BALB/c mice. Subsequently, a periodontal defect was created beside the maxillary first molar and filled with ceramic bovine bone. Finally, the chimeric mice were divided into three groups and were observed 3, 14 and 28 days later respectively. The involvement of BMSCs in periodontal defects was analysed using an in vivo imaging system and immunohistochemical staining of CD45, CD105 and CD31. Cell surface marker expression in injured tissue was also compared with that in normal tissue. RESULTS: Increasing numbers of BMSCs migrated into the periodontal defect with time. The distribution was initially limited to ceramic bovine bone and then around blood vessels and near alveolar bone. Furthermore, expression of CD105 and CD31 was much higher in injured periodontal tissue than that in healthy periodontium, although CD45 was not expressed in either of these tissues. CONCLUSION: BMSCs, but not haemopoietic stem cells, were involved in periodontal defect; they entered the periodontium probably via blood vessels.


Subject(s)
Alveolar Bone Loss/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Alveolar Bone Loss/pathology , Alveolar Process/pathology , Animals , Antigens, CD/analysis , Bone Substitutes/therapeutic use , Cattle , Cell Movement/physiology , Endoglin , Intracellular Signaling Peptides and Proteins/analysis , Leukocyte Common Antigens/analysis , Luminescent Agents , Luminescent Proteins , Male , Mice , Mice, Inbred BALB C , Models, Animal , Periodontal Ligament/pathology , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Protein Isoforms/analysis , Time Factors , Transplantation Chimera/physiology , Whole Body Imaging/methods , Wound Healing/physiology , Red Fluorescent Protein
14.
Br J Haematol ; 166(4): 557-65, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24779895

ABSTRACT

This study aimed to describe kinetics of complete donor chimerism occurrence (cDC, >99·9% donor) after unrelated cord blood transplantation (UCBT), to identify its predictive factors and its impact on post-transplant outcome. Ninety-four children who received single UCBT after a myeloablative conditioning regimen had blood chimerism evaluation at predefined post-transplant dates, using a real-time polymerase chain reaction method with 0·1% sensitivity. Cumulative incidence of cDC at 1 year post-transplantation was 61·8%. Three predictive factors were identified in multivariate analysis: history of malignant disease (P = 0·03), older age (above 2·16 years, the first quartile of age, P = 0·0055) and higher level of cord/recipient human leucocyte antigen mismatch (4/6 vs. 5-6/6, P < 0·001) increased the probability of post-transplant cDC. Although graft cell dose had a strong impact on haematological recovery, it did not apparently influence cDC occurrence. Early cDC (i.e. more than 99·9% donor chimerism on days 15-30 post-transplant) appeared useful to predict engraftment (P = 0·003) as well as acute and chronic graft-versus-host disease (GvHD). Severe acute or chronic GvHD never occurred in patients with DC ≤99·9%, suggesting than even minimal residual host haematopoiesis is associated with a very low risk of GvHD after UCBT.


Subject(s)
Cord Blood Stem Cell Transplantation/adverse effects , Transplantation Chimera/physiology , Adolescent , Allografts/physiology , Child , Child, Preschool , Female , Graft vs Host Disease/etiology , Hematopoiesis/physiology , Humans , Infant , Male , Minor Histocompatibility Antigens , Prospective Studies , Transplantation Conditioning/methods , Transplantation, Homologous , Treatment Outcome
15.
Lancet ; 383(9915): 436-48, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24161820

ABSTRACT

BACKGROUND: In chronic granulomatous disease allogeneic haemopoietic stem-cell transplantation (HSCT) in adolescents and young adults and patients with high-risk disease is complicated by graft-failure, graft-versus-host disease (GVHD), and transplant-related mortality. We examined the effect of a reduced-intensity conditioning regimen designed to enhance myeloid engraftment and reduce organ toxicity in these patients. METHODS: This prospective study was done at 16 centres in ten countries worldwide. Patients aged 0-40 years with chronic granulomatous disease were assessed and enrolled at the discretion of individual centres. Reduced-intensity conditioning consisted of high-dose fludarabine (30 mg/m(2) [infants <9 kg 1·2 mg/kg]; one dose per day on days -8 to -3), serotherapy (anti-thymocyte globulin [10 mg/kg, one dose per day on days -4 to -1; or thymoglobuline 2·5 mg/kg, one dose per day on days -5 to -3]; or low-dose alemtuzumab [<1 mg/kg on days -8 to -6]), and low-dose (50-72% of myeloablative dose) or targeted busulfan administration (recommended cumulative area under the curve: 45-65 mg/L × h). Busulfan was administered mainly intravenously and exceptionally orally from days -5 to -3. Intravenous busulfan was dosed according to weight-based recommendations and was administered in most centres (ten) twice daily over 4 h. Unmanipulated bone marrow or peripheral blood stem cells from HLA-matched related-donors or HLA-9/10 or HLA-10/10 matched unrelated-donors were infused. The primary endpoints were overall survival and event-free survival (EFS), probabilities of overall survival and EFS at 2 years, incidence of acute and chronic GVHD, achievement of at least 90% myeloid donor chimerism, and incidence of graft failure after at least 6 months of follow-up. FINDINGS: 56 patients (median age 12·7 years; IQR 6·8-17·3) with chronic granulomatous disease were enrolled from June 15, 2003, to Dec 15, 2012. 42 patients (75%) had high-risk features (ie, intractable infections and autoinflammation), 25 (45%) were adolescents and young adults (age 14-39 years). 21 HLA-matched related-donor and 35 HLA-matched unrelated-donor transplants were done. Median time to engraftment was 19 days (IQR 16-22) for neutrophils and 21 days (IQR 16-25) for platelets. At median follow-up of 21 months (IQR 13-35) overall survival was 93% (52 of 56) and EFS was 89% (50 of 56). The 2-year probability of overall survival was 96% (95% CI 86·46-99·09) and of EFS was 91% (79·78-96·17). Graft-failure occurred in 5% (three of 56) of patients. The cumulative incidence of acute GVHD of grade III-IV was 4% (two of 56) and of chronic graft-versus-host disease was 7% (four of 56). Stable (≥90%) myeloid donor chimerism was documented in 52 (93%) surviving patients. INTERPRETATION: This reduced-intensity conditioning regimen is safe and efficacious in high-risk patients with chronic granulomatous disease. FUNDING: None.


Subject(s)
Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Adolescent , Adult , Alemtuzumab , Antibodies, Monoclonal, Humanized/administration & dosage , Antilymphocyte Serum/administration & dosage , Busulfan/administration & dosage , Child , Child, Preschool , Drug Therapy, Combination , Graft Survival/drug effects , Graft vs Host Disease/prevention & control , HLA Antigens , Humans , Immunosuppressive Agents/administration & dosage , Infant , Prospective Studies , Transplantation Chimera/physiology , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Young Adult
16.
J Reprod Dev ; 59(6): 580-7, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24077020

ABSTRACT

The Japanese quail (Coturnix japonica) is a valuable bird as both an experimental animal, for a wide range of scientific disciplines, and an agricultural animal, for the production of eggs and meat. Cryopreservation of PGCs would be a feasible strategy for the conservation of both male and female fertility cells in Japanese quail. However, the effects of freeze-thaw treatment on viability, migration ability and germline transmission ability of quail PGCs still remain unclear. In the present study, male and female PGCs were isolated from the blood of 2-day-old embryos, which were cooled by slow freezing and then cryopreserved at -196 C for 77-185 days, respectively. The average recovery rate of PGCs after freeze-thawing was 47.0%. The viability of PGCs in the frozen group was significantly lower than that of the control group (P<0.05) (85.5% vs. 95.1%). Both fresh and Frozen-thawed PGCs that were intravascularly transplanted into recipient embryos migrated toward and were incorporated into recipient gonads, although the number of PGCs settled in the gonads was 48.5% lower in the frozen group than in the unfrozen control group (P<0.05). Genetic cross analysis revealed that one female and two male recipients produced live progeny derived from the frozen-thawed PGCs. The frequency of donor-derived offspring was slightly lower than that of unfrozen controls, but the difference was not significant (4.0 vs. 14.0%). These results revealed that freeze-thaw treatment causes a decrease in viability, migration ability and germline transmission ability of PGCs in quail.


Subject(s)
Coturnix/physiology , Cryopreservation/veterinary , Embryonic Stem Cells/physiology , Endangered Species , Ovum/physiology , Spermatozoa/physiology , Transplantation Chimera/physiology , Animals , Biological Specimen Banks , Cell Movement , Cell Survival , Coturnix/embryology , Coturnix/genetics , Down-Regulation , Embryo Culture Techniques/veterinary , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Feasibility Studies , Female , Fertility , Japan , Male , Mutation , Ovum/cytology , Ovum/transplantation , Spermatozoa/cytology , Spermatozoa/transplantation , Stem Cell Transplantation , Transplantation Chimera/genetics
17.
Blood ; 122(9): 1669-77, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23869083

ABSTRACT

Induction of mixed hematopoietic chimerism results in donor-specific immunological tolerance by apoptosis-mediated deletion of donor-reactive lymphocytes. A broad clinical application of this approach is currently hampered by limited predictability and toxicity of the available conditioning protocols. We developed a new therapeutic approach to induce mixed chimerism and tolerance by a direct pharmacological modulation of the intrinsic apoptosis pathway in peripheral T cells. The proapoptotic small-molecule Bcl-2 inhibitor ABT-737 promoted mixed chimerism induction and reversed the antitolerogenic effect of calcineurin inhibitors by boosting the critical role of the proapoptotic Bcl-2 factor Bim. A short conditioning protocol with ABT-737 in combination with costimulation blockade and low-dose cyclosporine A resulted in a complete deletion of peripheral donor-reactive lymphocytes and was sufficient to induce mixed chimerism and robust systemic tolerance across full major histocompatibility complex barriers, without myelosuppression and by using moderate doses of bone marrow cells. Thus, immunological tolerance can be achieved by direct modulation of the intrinsic apoptosis pathway in peripheral lymphocytes-a new approach to translate immunological tolerance into clinically applicable protocols.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Graft Survival/drug effects , Hematopoiesis/drug effects , Molecular Targeted Therapy/methods , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Transplantation Chimera , Animals , Apoptosis/immunology , Biphenyl Compounds/therapeutic use , Cells, Cultured , Graft Survival/physiology , Hematopoiesis/physiology , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Nitrophenols/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/therapeutic use , Transplantation Chimera/immunology , Transplantation Chimera/physiology , Transplantation Conditioning/methods , Transplantation Tolerance/drug effects , Up-Regulation/drug effects , Up-Regulation/immunology
18.
Poult Sci ; 92(4): 1073-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23472030

ABSTRACT

Ovaries from Rhode Island Red donors were transplanted orthotopically into White Leghorn recipients. At maturation, recipients were mated with Rhode Island Red roosters to test the origin of their ovaries, using plumage coloration as a marker. A chick with chimeric plumage coloration was produced, indicating mechanisms that produce follicles with both donor and recipient ovarian contents. This study suggests that ovarian transplantation could be useful for in vivo studies of cytological and molecular mechanisms involved in avian folliculogenesis.


Subject(s)
Chickens , Ovary/transplantation , Pigmentation , Transplantation Chimera/growth & development , Transplantation Chimera/genetics , Animals , Feathers/physiology , Female , Transplantation Chimera/physiology
19.
Hepatology ; 57(3): 1215-24, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23080021

ABSTRACT

UNLABELLED: The potential lineage relationship between hepatic oval cells, small hepatocyte-like progenitor cells (SHPCs), and hepatocytes in liver regeneration is debated. To test whether mature hepatocytes can give rise to SHPCs, rats with dipeptidyl peptidase IV (DPPIV) chimeric livers, which harbored endogenous DPPIV-deficient hepatocytes and transplanted DPPIV-positive hepatocytes, were subjected to retrorsine treatment followed by partial hepatectomy (PH). DPPIV-positive hepatocytes comprised about half of the DPPIV chimeric liver mass. Tissues from DPPIV chimeric livers after retrorsine/PH treatment showed large numbers of SHPC clusters. None of the SHPC clusters were stained positive for DPPIV in any analyzed samples. Furthermore, serial sections stained for gamma-glutamyl-transpeptidase (GGT, a marker of fetal hepatoblasts) and glucose-6-phosphatase (G6Pase, a marker of mature hepatocytes) showed inverse expression of the two enzymes and a staining pattern consistent with a lineage that begins with GGT(+)/G6Pase(-) to GGT(-)/G6Pase(+) within a single SHPC cluster. Using double immunofluorescence staining for markers specific for hepatic oval cells and hepatocytes in serial sections, oval cell proliferations with CK-19(+)/laminin(+) and OV-6(+)/C/EBP-α(-) were shown to extend from periportal areas into the SPHC clusters, differentiating into hepatic lineage by progressive loss of CK-19/laminin expression and appearance of C/EBP-α expression towards the cluster side. Cells in the epithelial cell adhesion molecule (EpCAM(+)) SHPC clusters showed membranous EpCAM(+)/HNF-4α(+) (hepatocyte nuclear factor-4α) staining and were contiguous to the surrounding cytoplasmic EpCAM(+)/HNF-4α(-) ductular oval cells. Extensive elimination of oval cell response by repeated administration of 4,4'-methylenedianiline (DAPM) to retrorsine-exposed rats impaired the emergence of SHPC clusters. CONCLUSION: These findings highly suggest the hepatic oval cells but not mature hepatocytes as the origin of SHPC clusters in retrorsine-exposed rats.


Subject(s)
Hepatocytes/cytology , Hepatocytes/physiology , Pyrrolizidine Alkaloids/pharmacology , Stem Cells/cytology , Stem Cells/physiology , Transplantation Chimera/physiology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Lineage/physiology , Cell Size , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Female , Hepatectomy/methods , Hepatocytes/drug effects , Male , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Rats , Rats, Inbred F344 , Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL