Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
J Am Soc Nephrol ; 33(2): 357-373, 2022 02.
Article in English | MEDLINE | ID: mdl-35046131

ABSTRACT

BACKGROUND: Receptor-interacting protein kinase 3 (RIPK3), a component of necroptosis pathways, may have an independent role in inflammation. It has been unclear which RIPK3-expressing cells are responsible for the anti-inflammatory effect of overall Ripk3 deficiency and whether Ripk3 deficiency protects against kidney inflammation occurring in the absence of tubular cell death. METHODS: We used chimeric mice with bone marrow from wild-type and Ripk3-knockout mice to explore RIPK3's contribution to kidney inflammation in the presence of folic acid-induced acute kidney injury AKI (FA-AKI) or absence of AKI and kidney cell death (as seen in systemic administration of the cytokine TNF-like weak inducer of apoptosis [TWEAK]). RESULTS: Tubular and interstitial cell RIPK3 expressions were increased in murine AKI. Ripk3 deficiency decreased NF-κB activation and kidney inflammation in FA-AKI but did not prevent kidney failure. In the chimeric mice, RIPK3-expressing bone marrow-derived cells were required for early inflammation in FA-AKI. The NLRP3 inflammasome was not involved in RIPK3's proinflammatory effect. Systemic TWEAK administration induced kidney inflammation in wild-type but not Ripk3-deficient mice. In cell cultures, TWEAK increased RIPK3 expression in bone marrow-derived macrophages and tubular cells. RIPK3 mediated TWEAK-induced NF-κB activation and inflammatory responses in bone marrow-derived macrophages and dendritic cells and in Jurkat T cells; however, in tubular cells, RIPK3 mediated only TWEAK-induced Il-6 expression. Furthermore, conditioned media from TWEAK-exposed wild-type macrophages, but not from Ripk3-deficient macrophages, promoted proinflammatory responses in cultured tubular cells. CONCLUSIONS: RIPK3 mediates kidney inflammation independently from tubular cell death. Specific targeting of bone marrow-derived RIPK3 may limit kidney inflammation without the potential adverse effects of systemic RIPK3 targeting.


Subject(s)
Acute Kidney Injury/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Bone Marrow/metabolism , Cytokine TWEAK/administration & dosage , Disease Models, Animal , Folic Acid/toxicity , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Jurkat Cells , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Transplantation Chimera/metabolism , Up-Regulation
2.
Stem Cell Reports ; 16(11): 2577-2588, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34678209

ABSTRACT

A reliable source of human hepatocytes and transplantable livers is needed. Interspecies embryo complementation, which involves implanting donor human stem cells into early morula/blastocyst stage animal embryos, is an emerging solution to the shortage of transplantable livers. We review proposed mutations in the recipient embryo to disable hepatogenesis, and discuss the advantages of using fumarylacetoacetate hydrolase knockouts and other genetic modifications to disable hepatogenesis. Interspecies blastocyst complementation using porcine recipients for primate donors has been achieved, although percentages of chimerism remain persistently low. Recent investigation into the dynamic transcriptomes of pigs and primates have created new opportunities to intimately match the stage of developing animal embryos with one of the many varieties of human induced pluripotent stem cell. We discuss techniques for decreasing donor cell apoptosis, targeting donor tissue to endodermal structures to avoid neural or germline chimerism, and decreasing the immunogenicity of chimeric organs by generating donor endothelium.


Subject(s)
Gene Editing/methods , Hydrolases/genetics , Liver Transplantation/methods , Living Donors , Transplantation Chimera/genetics , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Profiling/methods , Humans , Hydrolases/metabolism , Induced Pluripotent Stem Cells/metabolism , Transplantation Chimera/metabolism
3.
Sci Rep ; 10(1): 13315, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32769989

ABSTRACT

Using human induced pluripotent stem cells (iPSC), recent studies have shown that the events underlying autism spectrum disorders (ASD) can occur during neonatal development. We previously analyzed the iPSC-derived pyramidal cortical neurons of a subset of patients with ASD carrying de novo heterozygous mutations in postsynaptic SHANK3 protein, in culture. We reported altered spinogenesis of those neurons. The transplantation of human iPSC-derived neuronal precursors into mouse brain represents a novel option for in vivo analysis of mutations affecting the human brain. In this study, we transplanted the neuronal precursor cells (NPC) into the cortex of newborn mice to analyze their integration and maturation at early stages of development and studied axonal projections of transplanted human neurons into adult mouse brain. We then co-transplanted NPC from a control individual and from a patient carrying a de novo heterozygous SHANK3 mutation. We observed a reduction in cell soma size of selective neuronal categories and in axonal projections at 30 days post-transplantation. In contrast to previous in vitro studies, we did not observe any alteration in spinogenesis at this early age. The humanized chimeric mouse models offer the means to analyze ASD-associated mutations further and provide the opportunity to visualize phenotypes in vivo.


Subject(s)
Autism Spectrum Disorder/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation , Nerve Tissue Proteins/metabolism , Neural Stem Cells , Pyramidal Cells/metabolism , Stem Cell Transplantation , Transplantation Chimera/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Cell Line , Disease Models, Animal , Heterografts , Humans , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Pyramidal Cells/pathology , Transplantation Chimera/genetics
4.
Drug Metab Dispos ; 48(10): 934-943, 2020 10.
Article in English | MEDLINE | ID: mdl-32665417

ABSTRACT

The PXB-mouse is potentially a useful in vivo model to predict human hepatic metabolism and clearance. Four model compounds, [14C]desloratadine, [3H]mianserin, cyproheptadine, and [3H]carbazeran, all reported with disproportionate human metabolites, were orally administered to PXB- or control SCID mice to elucidate the biotransformation of each of them. For [14C]desloratadine in PXB-mice, O-glucuronide of 3-hydroxydesloratadine was observed as the predominant metabolite in both the plasma and urine. Both 3-hydroxydesloratadine and its O-glucuronide were detected as major drug-related materials in the bile, whereas only 3-hydroxydesloratadine was detected in the feces, suggesting that a fraction of 3-hydroxydesloratadine in feces was derived from deconjugation of its O-glucuronide by gut microflora. This information can help understand the biliary clearance mechanism of a drug and may fill the gap in a human absorption, distribution, metabolism, and excretion study, in which the bile samples are typically not available. The metabolic profiles in PXB-mice were qualitatively similar to those reported in humans in a clinical study in which 3-hydroxydesloratadine and its O-glucuronide were major and disproportionate metabolites compared with rat, mouse, and monkey. In the control SCID mice, neither of the metabolites was detected in any matrix. Similarly, for the other three compounds, all human specific or disproportionate metabolites were detected at a high level in PXB-mice, but they were either minimally observed or not observed in the control mice. Data from these four compounds indicate that studies in PXB-mice can help predict the potential for the presence of human disproportionate metabolites (relative to preclinical species) prior to conducting clinical studies and understand the biliary clearance mechanism of a drug. SIGNIFICANCE STATEMENT: Studies in PXB-mice have successfully predicted the human major and disproportionate metabolites compared with preclinical safety species for desloratadine, mianserin, cyproheptadine, and carbazeran. In addition, biliary excretion data from PXB-mice can help illustrate the human biliary clearance mechanism of a drug.


Subject(s)
Hepatobiliary Elimination , Liver/metabolism , Animals , Bile/metabolism , Biotransformation , Carbamates/administration & dosage , Carbamates/pharmacokinetics , Cyproheptadine/administration & dosage , Cyproheptadine/pharmacokinetics , Drug Evaluation, Preclinical/methods , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Liver/cytology , Loratadine/administration & dosage , Loratadine/analogs & derivatives , Loratadine/pharmacokinetics , Male , Mianserin/administration & dosage , Mianserin/pharmacokinetics , Mice , Transplantation Chimera/metabolism
5.
Drug Metab Dispos ; 48(7): 580-586, 2020 07.
Article in English | MEDLINE | ID: mdl-32357972

ABSTRACT

Carbazeran is a potent phosphodiesterase inhibitor with species-dependent metabolic profiles in rats, dogs, and humans. In this study, we investigated the aldehyde oxidase (AOX)-mediated oxidation of carbazeran to 4-oxo derivatives in chimeric NOD/Shi-scid IL2 receptor gamma-null mice expressing a herpes simplex virus type 1 thymidine kinase transgene with humanized livers (humanized-liver mice). Liver cytosolic fractions from humanized-liver mouse effectively catalyzed carbazeran 4-oxidation with high affinity for the substrate, similar to those of the human liver cytosolic fractions and recombinant human AOX1 protein. Furthermore, hepatocytes prepared from humanized-liver mice and humans also exhibited substantial metabolism via carbazeran 4-oxidation. After a single oral administration of carbazeran (10 mg/kg), plasma levels of 4-oxo-carbazeran, N-desethyl-4-oxo-carbazeran, and 6,7-dimethoxy-1-[4-(hydroxy)-piperidino]-4-phthalazinone (three human metabolites formed via 4-oxidation) were higher in humanized-liver mice than in the control mice. In contrast, plasma levels of O-desmethylcarbazeran (a major metabolite in dogs) in control mice were higher than those in the humanized-liver mice. Relative excreted amounts of the three 4-oxidation-derived human-specific metabolites in the urine and feces were greater for humanized-liver mice than control mice, whereas the relative excreted amounts of O-desmethylcarbazeran were predominant in the urine and feces of control mice. Thus, the production of carbazeran 4-oxo derivatives was elevated in humanized-liver mice compared with control mice, in agreement with our in vitro enzyme-mediated oxidation data. These results suggest that hepatic human AOX1 functions in humanized-liver mice at the in vivo level and that humanized-liver mice may be useful for predicting drug metabolism in humans, at least with regard to human AOX1-dependent metabolism. SIGNIFICANCE STATEMENT: We found that the production of carbazeran 4-oxo derivatives was higher in humanized-liver mice than in control mice. These results were supported by the fact that carbazeran was rapidly metabolized to 4-oxo-carbazeran in humanized-liver mouse hepatocytes expressing human aldehyde oxidase 1. These results suggest that human aldehyde oxidase 1 is functional in humanized-liver mice in vivo and that chimeric NOD/Shi-scid IL2 receptor gamma-null mice expressing a herpes simplex virus type 1 thymidine kinase transgene transplanted with human hepatocytes may be a suitable model animal for predicting aldehyde oxidase-dependent biotransformation of drugs in humans.


Subject(s)
Aldehyde Oxidase/metabolism , Carbamates/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Animals , Biotransformation , Carbamates/administration & dosage , Cells, Cultured , Child , Child, Preschool , Dogs , Drug Evaluation, Preclinical/methods , Enzyme Assays , Feasibility Studies , Female , Guinea Pigs , Hepatocytes/metabolism , Humans , Interleukin Receptor Common gamma Subunit/genetics , Macaca fascicularis , Male , Mice , Mice, Knockout , Middle Aged , Oxidation-Reduction , Rabbits , Rats , Recombinant Proteins/metabolism , Swine , Swine, Miniature , Transplantation Chimera/metabolism , Young Adult
6.
Chem Res Toxicol ; 33(2): 634-639, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31854189

ABSTRACT

To investigate the respective roles of cytochromes P450 2C9 and 3A in drug oxidation in human livers, the in vivo pharmacokinetics of S-warfarin and diclofenac were analyzed after intravenous administrations in chimeric mice that had been transplanted with human hepatocytes. P450 2C9 was metabolically inactivated in the humanized mice by orally pretreating them with tienilic acid. After intravenous administration of S-warfarin, a significant difference in the concentration-time profiles of the primary metabolite 7-hydroxywarfarin between untreated mice and mice treated with tienilic acid was observed. In contrast, there were no apparent differences in the profiles for S-warfarin between the treated and untreated groups. The mean values of the maximum concentrations (Cmax) and the areas under the plasma concentration versus time curves (AUCinfinity) for 7-hydroxywarfarin were significantly lower (22 and 16% of the untreated values, respectively) in the treated group. This presumably resulted from suppressed P450 2C9 activity in the primary oxidative metabolism in vivo in the treated group. After diclofenac administration, plasma levels of diclofenac, 5-hydroxydiclofenac, and diclofenac acylglucuronide were roughly similar in pretreated and untreated mice. However, the mean Cmax and AUCinfinity values for 4'-hydroxydiclofenac were significantly lower (38 and 53% of the untreated group, respectively) in the treated group. The reported value of ∼0.8 for the fraction of S-warfarin metabolized to 7-hydroxywarfarin mediated by P450 2C9 in in vitro systems was similar to the value implied by the present humanized-liver mouse model pretreated with tienilic acid in which the AUC of 7-hydroxywarfarin was reduced by 84%. In contrast, the fractions of diclofenac metabolized to 4'-hydroxydiclofenac in in vitro and in vivo experiments were inconsistent. These results suggested that humanized-liver mice orally treated with tienilic acid might constitute an in vivo model for metabolically inactivated P450 2C9 in human hepatocytes transplanted into chimeric mice. Moreover, diclofenac, a typical in vitro P450 2C9 probe substrate, was cleared differently in vitro and in humanized-liver mice in vivo.


Subject(s)
Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Diclofenac/analogs & derivatives , Hepatocytes/metabolism , Transplantation Chimera/metabolism , Animals , Diclofenac/metabolism , Humans , Hydroxylation , Mice
7.
J Clin Invest ; 129(8): 3293-3309, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31135379

ABSTRACT

Idiopathic Pulmonary Fibrosis (IPF) is a deadly disease with limited therapies. Tissue fibrosis is associated with Type 2 immune response, although the causal contribution of immune cells is not defined. The AP-1 transcription factor Fra-2 is upregulated in IPF lung sections and Fra-2 transgenic mice (Fra-2tg) exhibit spontaneous lung fibrosis. Here we show that Bleomycin-induced lung fibrosis is attenuated upon myeloid-inactivation of Fra-2 and aggravated in Fra-2tg bone marrow chimeras. Type VI collagen (ColVI), a Fra-2 transcriptional target, is up-regulated in three lung fibrosis models, and macrophages promote myofibroblast activation in vitro in a ColVI- and Fra-2-dependent manner. Fra-2 or ColVI inactivation does not affect macrophage recruitment and alternative activation, suggesting that Fra-2/ColVI specifically controls the paracrine pro-fibrotic activity of macrophages. Importantly, ColVI knock-out mice (KO) and ColVI-KO bone marrow chimeras are protected from Bleomycin-induced lung fibrosis. Therapeutic administration of a Fra-2/AP-1 inhibitor reduces ColVI expression and ameliorates fibrosis in Fra-2tg mice and in the Bleomycin model. Finally, Fra-2 and ColVI positively correlate in IPF patient samples and co-localize in lung macrophages. Therefore, the Fra-2/ColVI pro-fibrotic axis is a promising biomarker and therapeutic target for lung fibrosis, and possibly other fibrotic diseases.


Subject(s)
Fos-Related Antigen-2/biosynthesis , Idiopathic Pulmonary Fibrosis/metabolism , Macrophages/immunology , Myofibroblasts/metabolism , Allografts , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Transplantation , Collagen Type VI/biosynthesis , Collagen Type VI/genetics , Fos-Related Antigen-2/genetics , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Macrophages/pathology , Mice , Mice, Knockout , Myofibroblasts/pathology , Transplantation Chimera/genetics , Transplantation Chimera/metabolism
8.
Curr Protoc Immunol ; 123(1): e56, 2018 11.
Article in English | MEDLINE | ID: mdl-30222250

ABSTRACT

Bone marrow transplantation is the standard of care for a host of diseases such as leukemia and multiple myeloma, as well as genetically inherited metabolic diseases affecting the central nervous system. In mouse models, bone marrow transplantation has proven a valuable tool for understanding the hematopoietic system and the homing of hematopoietic cells to their target organs. Many techniques have been developed to create chimeric mice, animals with a hematopoietic system derived from a genetic background that differs from the rest of the body. Current genetic tools allow for virtually limitless possibilities in the choice of donor mice. This protocol describes methods of bone marrow transplantation in mouse models for studies of the brain under basal and pathological conditions. Specific points to be addressed include the preparation of recipient mice by irradiation or chemotherapy; the choice, isolation, and injection of donor cells; and analytical methods such as fluorescence-activated cell sorting and immunostaining. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Bone Marrow Transplantation , Brain Diseases/metabolism , Brain/metabolism , Transplantation Chimera/metabolism , Animals , Brain/pathology , Brain Diseases/pathology , Brain Diseases/therapy , Disease Models, Animal , Graft Survival , Inflammation/metabolism , Inflammation/pathology
9.
J Pediatr Hematol Oncol ; 40(8): e486-e489, 2018 11.
Article in English | MEDLINE | ID: mdl-30188352

ABSTRACT

The mesenchymal stem cell (MSC) role after allogeneic hematopoietic stem cell transplantation (HSCT) is still a matter of debate; in particular, MSC engraftment in recipient bone marrow (BM) is unclear. A total of 46 patients were analyzed for MSC and hemopoietic stem cell engraftment after HSCT. The majority of patients had the BM as the stem cell source, and acute leukemia was the main indication for HSCT. Mesenchymal and hematopoietic stem cell chimerism analysis was carried out through specific polymorphic tandemly repeated regions. All patients reached complete donor engraftment; no evidence of donor-derived MSC engraftment was noted. Our data indicate that MSCs after HSCT remain of recipient origin despite the following: (i) myeloablative conditioning; (ii) the stem cell source; (iii) the interval from HSCT to BM analysis; (iv) the underlying disease before HSCT; and (v) the patients' or the donors' age at HSCT.


Subject(s)
Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Transplantation Chimera/metabolism , Adolescent , Adult , Aged , Allografts , Child , Child, Preschool , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
10.
PLoS One ; 13(5): e0197604, 2018.
Article in English | MEDLINE | ID: mdl-29782549

ABSTRACT

Idiopathic pulmonary fibrosis is a devastating disease with poor prognosis. The pathogenic role of the lysophospholipid mediator sphingosine-1-phosphate and its receptor S1PR2 in lung fibrosis is unknown. We show here that genetic deletion of S1pr2 strikingly attenuated lung fibrosis induced by repeated injections of bleomycin in mice. We observed by using S1pr2LacZ/+ mice that S1PR2 was expressed in alveolar macrophages, vascular endothelial cells and alveolar epithelial cells in the lung and that S1PR2-expressing cells accumulated in the fibrotic legions. Bone marrow chimera experiments suggested that S1PR2 in bone marrow-derived cells contributes to the development of lung fibrosis. Depletion of macrophages greatly attenuated lung fibrosis. Bleomycin administration stimulated the mRNA expression of the profibrotic cytokines IL-13 and IL-4 and the M2 markers including arginase 1, Fizz1/Retnla, Ccl17 and Ccl24 in cells collected from broncho-alveolar lavage fluids (BALF), and S1pr2 deletion markedly diminished the stimulated expression of these genes. BALF cells from bleomycin-administered wild-type mice showed a marked increase in phosphorylation of STAT6, a transcription factor which is activated downstream of IL-13, compared with saline-administered wild-type mice. Interestingly, in bleomycin-administered S1pr2-/- mice, STAT6 phosphorylation in BALF cells was substantially diminished compared with wild-type mice. Finally, pharmacological S1PR2 blockade in S1pr2+/+ mice alleviated bleomycin-induced lung fibrosis. Thus, S1PR2 facilitates lung fibrosis through the mechanisms involving augmentation of IL-13 expression and its signaling in BALF cells, and represents a novel target for treating lung fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/etiology , Interleukin-13/metabolism , Macrophages/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Interleukin-13/genetics , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction , Sphingosine-1-Phosphate Receptors , Transplantation Chimera/genetics , Transplantation Chimera/metabolism , Up-Regulation
11.
J Clin Invest ; 128(8): 3312-3318, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29715199

ABSTRACT

For gene therapy of gain-of-function autosomal dominant diseases, either correcting or deleting the disease allele is potentially curative. To test whether there may be an advantage of one approach over the other for WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome - a primary immunodeficiency disorder caused by gain-of-function autosomal dominant mutations in chemokine receptor CXCR4 - we performed competitive transplantation experiments using both lethally irradiated WT (Cxcr4+/+) and unconditioned WHIM (Cxcr4+/w) recipient mice. In both models, hematopoietic reconstitution was markedly superior using BM cells from donors hemizygous for Cxcr4 (Cxcr4+/o) compared with BM cells from Cxcr4+/+ donors. Remarkably, only approximately 6% Cxcr4+/o hematopoietic stem cell (HSC) chimerism after transplantation in unconditioned Cxcr4+/w recipient BM supported more than 70% long-term donor myeloid chimerism in blood and corrected myeloid cell deficiency in blood. Donor Cxcr4+/o HSCs differentiated normally and did not undergo exhaustion as late as 465 days after transplantation. Thus, disease allele deletion resulting in Cxcr4 haploinsufficiency was superior to disease allele repair in a mouse model of gene therapy for WHIM syndrome, allowing correction of leukopenia without recipient conditioning.


Subject(s)
Bone Marrow Transplantation , Haploinsufficiency , Immunologic Deficiency Syndromes , Leukopenia , Receptors, CXCR4 , Transplantation Chimera , Warts , Allografts , Animals , Disease Models, Animal , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Immunologic Deficiency Syndromes/therapy , Leukopenia/genetics , Leukopenia/metabolism , Leukopenia/pathology , Leukopenia/therapy , Mice , Mice, Mutant Strains , Primary Immunodeficiency Diseases , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Transplantation Chimera/genetics , Transplantation Chimera/metabolism , Warts/genetics , Warts/metabolism , Warts/pathology , Warts/therapy
12.
J Clin Invest ; 128(1): 219-232, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202467

ABSTRACT

Humoral rejection is the most common cause of solid organ transplant failure. Here, we evaluated a cohort of 49 patients who were successfully grafted with allogenic islets and determined that the appearance of donor-specific anti-HLA antibodies (DSAs) did not accelerate the rate of islet graft attrition, suggesting resistance to humoral rejection. Murine DSAs bound to allogeneic targets expressed by islet cells and induced their destruction in vitro; however, passive transfer of the same DSAs did not affect islet graft survival in murine models. Live imaging revealed that DSAs were sequestrated in the circulation of the recipients and failed to reach the endocrine cells of grafted islets. We used murine heart transplantation models to confirm that endothelial cells were the only accessible targets for DSAs, which induced the development of typical microvascular lesions in allogeneic transplants. In contrast, the vasculature of DSA-exposed allogeneic islet grafts was devoid of lesions because sprouting of recipient capillaries reestablished blood flow in grafted islets. Thus, we conclude that endothelial chimerism combined with vascular sequestration of DSAs protects islet grafts from humoral rejection. The reduced immunoglobulin concentrations in the interstitial tissue, confirmed in patients, may have important implications for biotherapies such as vaccines and monoclonal antibodies.


Subject(s)
Endothelium, Vascular/metabolism , Graft Rejection/metabolism , Graft Survival , Islets of Langerhans Transplantation , Isoantibodies/metabolism , Transplantation Chimera/metabolism , Allografts , Animals , Endothelium, Vascular/pathology , Female , Graft Rejection/pathology , Humans , Male , Mice
13.
Drug Metab Dispos ; 46(1): 11-19, 2018 01.
Article in English | MEDLINE | ID: mdl-29051147

ABSTRACT

The influence of transporters on the pharmacokinetics of drugs is being increasingly recognized, and DDIs via transporters may be a risk factor for adverse events. Cyclosporine A, a strong OATP inhibitor, has been reported to increase the systemic exposure of rosuvastatin, an OATP substrate, by 7.1-fold in clinical studies. PXB mice are chimeric mice with humanized livers that are highly repopulated with human hepatocytes and have been widely used for drug discovery in drug metabolism and pharmacokinetics studies. In the present study, we examined in vivo and in vitro DDIs between rosuvastatin and cyclosporine A in PXB mice and fresh human hepatocytes (PXB cells) obtained from PXB mice. We initially investigated the active transport of rosuvastatin into PXB cells, and found concentration-dependent uptake with a Michaelis-Menten constant value of 4.0 µmol/l and a Vmax value of 4.63 pmol/min per 106 cells. Cyclosporine A inhibited the uptake of rosuvastatin with an IC50 value of 0.21 µmol/l. We then examined in vivo DDIs, and the exposure of orally administered rosuvastatin increased by 3.3-fold and 11-fold in PXB mice pretreated with 10 and 50 mg/kg cyclosporine A, whereas it increased by 2.5-fold and 6.2-fold when rosuvastatin was administered intravenously, in studies that were conducted for considering gastrointestinal DDIs. The liver-to-blood concentration ratio of rosuvastatin was dose-dependently decreased by pretreatment with cyclosporine A in PXB mice and SCID mice. Observed DDIs in vivo were considered to be reasonable based on the estimated concentrations of cyclosporine A at the inlet to the liver and in the liver tissues of both mice. In conclusion, our results indicate that PXB mice might be a useful tool for predicting human OATP-mediated DDIs in drug discovery, and its limitation due to the differences of gastrointestinal condition from human should also be considered.


Subject(s)
Cyclosporine/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Organic Anion Transporters/metabolism , Rosuvastatin Calcium/pharmacology , Transplantation Chimera/metabolism , Animals , Drug Interactions , HEK293 Cells , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Liver/cytology , Liver/metabolism , Male , Mice , Mice, SCID , Models, Animal , Organic Anion Transporters/antagonists & inhibitors
14.
Blood ; 130(17): 1946-1948, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28887325

ABSTRACT

Novel curative therapies using genetic transfer of normal globin-producing genes into autologous hematopoietic stem cells (HSCs) are in clinical trials for patients with sickle cell disease (SCD). The percentage of transferred globin necessary to cure SCD is currently not known. In the setting of allogeneic nonmyeloablative HSC transplants (HSCTs), stable mixed chimerism is sufficient to reverse the disease. We regularly monitored 67 patients after HSCT. After initially robust engraftment, 3 of these patients experienced declining donor myeloid chimerism (DMC) levels with eventual return of disease. From this we discovered that 20% DMC is necessary to reverse the sickle phenotype. We subsequently developed a mathematical model to test the hypothesis that the percentage of DMC necessary is determined solely by differences between donor and recipient red blood cell (RBC) survival times. In our model, the required 20% DMC can be entirely explained by the large differences between donor and recipient RBC survival times. Our model predicts that the requisite DMC and therefore necessary level of transferred globin is lowest in patients with the highest reticulocyte counts and concomitantly shortened RBC lifespans.


Subject(s)
Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/therapy , Hematopoietic Stem Cell Transplantation , Myeloid Cells/pathology , Tissue Donors , Transplantation Chimera/metabolism , Adult , Hemoglobin, Sickle/metabolism , Homozygote , Humans , Phenotype , Transplantation, Homologous , Young Adult
15.
PLoS Biol ; 15(6): e2001930, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28632753

ABSTRACT

Superantigens (SAgs) are potent exotoxins secreted by Staphylococcus aureus and Streptococcus pyogenes. They target a large fraction of T cell pools to set in motion a "cytokine storm" with severe and sometimes life-threatening consequences typically encountered in toxic shock syndrome (TSS). Given the rapidity with which TSS develops, designing timely and truly targeted therapies for this syndrome requires identification of key mediators of the cytokine storm's initial wave. Equally important, early host responses to SAgs can be accompanied or followed by a state of immunosuppression, which in turn jeopardizes the host's ability to combat and clear infections. Unlike in mouse models, the mechanisms underlying SAg-associated immunosuppression in humans are ill-defined. In this work, we have identified a population of innate-like T cells, called mucosa-associated invariant T (MAIT) cells, as the most powerful source of pro-inflammatory cytokines after exposure to SAgs. We have utilized primary human peripheral blood and hepatic mononuclear cells, mouse MAIT hybridoma lines, HLA-DR4-transgenic mice, MAIThighHLA-DR4+ bone marrow chimeras, and humanized NOD-scid IL-2Rγnull mice to demonstrate for the first time that: i) mouse and human MAIT cells are hyperresponsive to SAgs, typified by staphylococcal enterotoxin B (SEB); ii) the human MAIT cell response to SEB is rapid and far greater in magnitude than that launched by unfractionated conventional T, invariant natural killer T (iNKT) or γδ T cells, and is characterized by production of interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-2, but not IL-17A; iii) high-affinity MHC class II interaction with SAgs, but not MHC-related protein 1 (MR1) participation, is required for MAIT cell activation; iv) MAIT cell responses to SEB can occur in a T cell receptor (TCR) Vß-specific manner but are largely contributed by IL-12 and IL-18; v) as MAIT cells are primed by SAgs, they also begin to develop a molecular signature consistent with exhaustion and failure to participate in antimicrobial defense. Accordingly, they upregulate lymphocyte-activation gene 3 (LAG-3), T cell immunoglobulin and mucin-3 (TIM-3), and/or programmed cell death-1 (PD-1), and acquire an anergic phenotype that interferes with their cognate function against Klebsiella pneumoniae and Escherichia coli; vi) MAIT cell hyperactivation and anergy co-utilize a signaling pathway that is governed by p38 and MEK1/2. Collectively, our findings demonstrate a pathogenic, rather than protective, role for MAIT cells during infection. Furthermore, we propose a novel mechanism of SAg-associated immunosuppression in humans. MAIT cells may therefore provide an attractive therapeutic target for the management of both early and late phases of severe SAg-mediated illnesses.


Subject(s)
Antigens, Bacterial/toxicity , Clonal Anergy , Models, Immunological , Mucosal-Associated Invariant T Cells/immunology , Staphylococcus aureus/immunology , Streptococcus pyogenes/immunology , Superantigens/toxicity , Animals , Antigens, Bacterial/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , Clonal Anergy/drug effects , Crosses, Genetic , Enterotoxins/metabolism , Enterotoxins/toxicity , Female , Humans , Hybridomas , Immunity, Innate , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Mucosal-Associated Invariant T Cells/cytology , Mucosal-Associated Invariant T Cells/drug effects , Mucosal-Associated Invariant T Cells/metabolism , Specific Pathogen-Free Organisms , Staphylococcus aureus/metabolism , Streptococcus pyogenes/metabolism , Superantigens/metabolism , Transplantation Chimera/blood , Transplantation Chimera/immunology , Transplantation Chimera/metabolism
16.
Xenotransplantation ; 24(3)2017 05.
Article in English | MEDLINE | ID: mdl-28397982

ABSTRACT

BACKGROUND: Blastocyst complementation is an important technique for generating chimeric organs in organ-deficient pigs, which holds great promise for solving the problem of a shortage of organs for human transplantation procedures. Porcine chimeras have been generated using embryonic germ cells, embryonic stem cells, and induced pluripotent stem cells; however, there are no authentic pluripotent stem cells for pigs. In previous studies, blastomeres from 4- to 8-cell-stage parthenogenetic embryos were able to generate chimeric fetuses efficiently, but the resulting fetuses did not produce live-born young. Here, we used early-stage embryos from somatic cell nuclear transfer (SCNT) to generate chimeric piglets by the aggregation method. Then, the distribution of chimerism in various tissues and organs was observed through the expression of enhanced green fluorescent protein (EGFP). METHODS: Initially, we determined whether 4- to 8- or 8- to 16-cell-stage embryos were more suitable to generate chimeric piglets. Chimeras were produced by aggregating two EGFP-tagged Wuzhishan minipig (WZSP) SCNT embryos and two Bama minipig (BMP) SCNT embryos. The chimeric piglets were identified by coat color and microsatellite and swine leukocyte antigen analyses. Moreover, the distribution of chimerism in various tissues and organs of the piglets was evaluated by EGFP expression. RESULTS: We found that more aggregated embryos were produced using 4- to 8-cell-stage embryos (157/657, 23.9%) than 8- to 16-cell-stage embryos (100/499, 20.0%). Thus, 4- to 8-cell-stage embryos were used for the generation of chimeras. The rate of blastocysts development after aggregating WZSP with BMP embryos was 50.6%. Transfer of 391 blastocysts developed from 4- to 8-cell-stage embryos to five recipients gave rise to 18 piglets, of which two (11.1%) were confirmed to be chimeric by their coat color and microsatellite examination of the skin. One of the chimeric piglets died at 35 days and was subsequently autopsied, whereas the other piglet was maintained for the following observations. The heart and kidneys of the dead piglet showed chimerism, whereas the spinal cord, stomach, pancreas, intestines, muscle, ovary, and brain had no chimerism. CONCLUSIONS: To our knowledge, this is the first report of porcine chimeras generated by aggregating 4- to 8-cell-stage blastomeres from SCNT. We detected chimerism only in the skin, heart, and kidneys. Collectively, these results indicate that aggregation using 4- to 8-cell-stage SCNT embryos offers a practical approach for producing chimeric minipigs. Furthermore, it also provides a potential platform for generating interspecific chimeras between pigs and non-human primates for xenotransplantation.


Subject(s)
Blastomeres/cytology , Nuclear Transfer Techniques , Swine, Miniature/embryology , Swine, Miniature/genetics , Transplantation Chimera/embryology , Transplantation Chimera/genetics , Animals , Animals, Genetically Modified , Cell Aggregation , Embryo Culture Techniques/methods , Embryo Transfer/methods , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histocompatibility Antigens Class I/genetics , Microsatellite Repeats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin Pigmentation/genetics , Swine , Transplantation Chimera/metabolism
17.
Eur J Drug Metab Pharmacokinet ; 42(6): 965-972, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28447323

ABSTRACT

BACKGROUND AND OBJECTIVE: Human in vitro and dog in vitro/in vivo researches indicate that the drug-drug interaction (DDI) of decreased plasma valproic acid (VPA) concentration by co-administration of carbapenem antibiotics is caused by inhibition of acylpeptide hydrolase (APEH)-mediated VPA acylglucuronide (VPA-G) hydrolysis by carbapenems. In this study, we investigated VPA disposition and APEH activities in TK-NOG chimeric mice, whose livers were highly replaced with human hepatocytes, to evaluate the utility of this animal model and the clinical relevance of the DDI mechanism. METHODS: VPA and VPA-G concentrations in plasma, urinary excretion of VPA-G and APEH activity in humanized livers were measured after co-administration of VPA with meropenem (MEPM) to chimeric mice. RESULTS: After co-administration with MEPM to the chimeric mice, plasma VPA concentration more rapidly decreased than without the co-administration. An increase in plasma AUC and urinary excretion of VPA-G was also observed. APEH activity in humanized livers was strongly inhibited even at 24 h after co-administration of MEPM to the chimeric mice. CONCLUSION: The DDI of VPA with carbapenems was successfully observed in chimeric mice with humanized livers. The DDI was caused by long-lasting inhibition of hepatic APEH-mediated VPA-G hydrolysis by carbapenems, which strongly supports the APEH-mediated mechanism of the clinical DDI. This is the first example showing the usefulness of chimeric mice with humanized livers for evaluation of a DDI via non-cytochrome P450 enzyme.


Subject(s)
Carbapenems/pharmacology , Liver/drug effects , Liver/metabolism , Valproic Acid/pharmacokinetics , Animals , Drug Interactions , Humans , Meropenem , Mice , Peptide Hydrolases/metabolism , Thienamycins/pharmacology , Transplantation Chimera/blood , Transplantation Chimera/metabolism , Transplantation Chimera/urine , Valproic Acid/blood , Valproic Acid/urine
18.
Tissue Eng Part A ; 22(9-10): 742-53, 2016 05.
Article in English | MEDLINE | ID: mdl-27068494

ABSTRACT

Cell-based therapies represent a promising alternative to orthotopic liver transplantation. However, therapeutic effects are limited by low cell engraftment rates. We recently introduced a technique creating human hepatocyte spheroids for potential therapeutic application. The aim of this study was to evaluate whether these spheroids are suitable for engraftment in diseased liver tissues. Intrasplenic spheroid transplantation into immunodeficient uPA/SCID/beige mice was performed. Hepatocyte transduction ability prior to transplantation was tested by lentiviral labeling using red-green-blue (RGB) marking. Eight weeks after transplantation, animals were sacrificed and livers were analyzed by immunohistochemistry and immunofluorescence. To investigate human hepatocyte-specific gene expression profiles in mice, quantitative real-time-PCR was applied. Human albumin and alpha-1-antitrypsin concentrations in mouse serum were quantified to assess the levels of human chimerism. Precultured human hepatocytes reestablished their physiological liver tissue architecture and function upon transplantation in mice. Positive immunohistochemical labeling of the proliferating cell nuclear antigen revealed that human hepatocytes retained their in vivo proliferation capacity. Expression profiles of human genes analyzed in chimeric mouse livers resembled levels determined in native human tissue. Extensive vascularization of human cell clusters was detected by demonstration of von Willebrand factor activity. To model gene therapy approaches, lentiviral transduction was performed ex vivo and fluorescent microscopic imaging revealed maintenance of RGB marking in vivo. Altogether, this is the first report demonstrating that cultured and retroviral transduced human hepatocyte spheroids are able to engraft and maintain their regenerative potential in vivo.


Subject(s)
Hepatocytes , Lentivirus , Liver/metabolism , Transduction, Genetic , Transplantation Chimera/metabolism , Animals , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hepatocytes/metabolism , Hepatocytes/transplantation , Heterografts , Humans , Mice , Mice, SCID , Mice, Transgenic , Transplantation Chimera/genetics
19.
Steroids ; 107: 139-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26774429

ABSTRACT

Anabolic androgenic steroids are prohibited by the World Anti-Doping Agency because of their adverse health and performance enhancing effects. Effective control of their misuse by detection in urine requires knowledge about their metabolism. In case of designer steroids, ethical objections limit the use of human volunteers to perform excretion studies. Therefore the suitability of alternative models needs to be investigated. In this study pooled human liver microsomes (HLM) and an uPA(+/+)-SCID chimeric mouse model were used to examine the metabolism of the designer steroid prostanozol as a reference standard. Metabolites were detected by GC-MS (full scan) and LC-MS/MS (precursor ion scan). In total twenty-four prostanozol metabolites were detected with the in vitro and in vivo metabolism studies, which could be grouped into two broad classes, those with a 17-hydroxy- and those with a 17-keto-substituent. Major first phase metabolic sites were tentatively identified as C-3'; C-4 and C-16. Moreover, 3'- and 16ß-hydroxy-17-ketoprostanozol could be unequivocally identified, since authentic reference material was available, in both models. Comparison with published data from humans showed a good correlation, except for phase II metabolism. As metabolites were in contrast to the human studies predominantly present in the free fraction. Two types of metabolites ((di)hydroxylated prostanozol metabolites) that have not been described before could be confirmed in a real positive doping control sample. Hence, the results provide further evidence for the applicability of chimeric mice and HLM to perform metabolism studies of designer steroids.


Subject(s)
Anabolic Agents , Microsomes, Liver/metabolism , Transplantation Chimera/metabolism , Anabolic Agents/pharmacokinetics , Anabolic Agents/pharmacology , Animals , Humans , Mice , Mice, SCID
20.
Bone Marrow Transplant ; 51(1): 127-31, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26437058

ABSTRACT

Central nervous system (CNS) complications have been described in patients undergoing allogeneic hematopoietic cell transplantation (alloHCT). Cerebrospinal fluid (CSF) analysis is included in the diagnostic workup in patients with neurological symptoms after alloHCT. CSF donor-recipient chimerism analysis usually is not used to evaluate patients with neurological complications after alloHCT. To assess the potential contribution of CSF donor-recipient chimerism in patients with neurological complications, we analyzed 85 CSF samples from 50 patients with neurological complications after alloHCT. After alloHCT, 21 patients showed the presence of recipient-derived DNA. In 13 of these patients, recurrence of the underlying disease was detected in CSF. There was a moderate correlation between the recipient DNA percentage as detected by short tandem repeat (STR) amplification and the cell concentration in CSF (Spearmann r: 0.66 P=0.004). The percentage of cells with immunophenotypic abnormalities from patients relapsing in the CNS detected by flow cytometry showed a strong correlation with the percentage of recipient-derived DNA in CSF assessed by STR analysis (Spearmann r: 0.83 P=0.0008). Donor-recipient chimerism analysis in CSF in patients with neurological symptoms after alloHCT is a practical, feasible and useful complementary method to the already established methodologies included in the diagnostic workup.


Subject(s)
Central Nervous System Diseases , Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Transplantation Chimera/metabolism , Adult , Aged , Allografts , Central Nervous System Diseases/cerebrospinal fluid , Central Nervous System Diseases/etiology , Central Nervous System Diseases/pathology , Female , Hematologic Neoplasms/cerebrospinal fluid , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL