Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Bioeng Transl Med ; 8(6): e10571, 2023 Nov.
Article En | MEDLINE | ID: mdl-38023726

Manufacture of chimeric antigen receptor (CAR)-T cells usually involves the use of viral delivery systems to achieve high transgene expression. However, it can be costly and may result in random integration of the CAR into the genome, creating several disadvantages including variation in transgene expression, functional gene silencing and potential oncogenic transformation. Here, we optimized the method of nonviral, CRISPR/Cas9 genome editing using large donor DNA delivery, knocked-in an anti-tumor single chain variable fragment (scFv) into the N-terminus of CD3ε and efficiently generated fusion protein (FP) T cells. These cells displayed FP integration within the TCR/CD3 complex, lower variability in gene expression compared to CAR-T cells and good cell expansion after transfection. CD3ε FP T cells were predominantly CD8+ effector memory T cells, and exhibited anti-tumor activity in vitro and in vivo. Dual targeting FP T cells were also generated through the incorporation of scFvs into other CD3 subunits and CD28. Compared to viral-based methods, this method serves as an alternative and versatile way of generating T cells with tumor-targeting receptors for cancer immunotherapy.

2.
Mol Ther Oncolytics ; 20: 325-341, 2021 Mar 26.
Article En | MEDLINE | ID: mdl-33614914

Chimeric antigen receptor (CAR) T cells have revolutionized blood cancer immunotherapy; however, their efficacy against solid tumors has been limited. A common mechanism of tumor escape from single target therapies is downregulation or mutational loss of the nominal epitope. Targeting multiple antigens may thus improve the effectiveness of CAR immunotherapies. We generated dual CAR-T cells targeting two tumor antigens: TAG-72 (tumor-associated glycoprotein 72) and CD47. TAG-72 is a pan-adenocarcinoma oncofetal antigen, highly expressed in ovarian cancers, with increased expression linked to disease progression. CD47 is ubiquitously overexpressed in multiple tumor types, including ovarian cancer; it is a macrophage "don't eat me" signal. However, CD47 is also expressed on many normal cells. To avoid this component of the dual CAR-T cells killing healthy tissue, we designed a truncated CD47 CAR devoid of intracellular signaling domains. The CD47 CAR facilitates binding to CD47+ cells, increasing the prospect of TAG-72+ cell elimination via the TAG-72 CAR. Furthermore, we could reduce the damage to normal tissue by monomerizing the CD47 CAR. Our results indicate that the co-expression of the TAG-72 CAR and the CD47-truncated monomer CAR on T cells could be an effective, dual CAR-T cell strategy for ovarian cancer, also applicable to other adenocarcinomas.

3.
Cell Rep ; 27(13): 3887-3901.e4, 2019 06 25.
Article En | MEDLINE | ID: mdl-31242421

A key feature of immune functional impairment with age is the progressive involution of thymic tissue responsible for naive T cell production. In this study, we identify two major phases of thymic epithelial cell (TEC) loss during aging: a block in mature TEC differentiation from the pool of immature precursors, occurring at the onset of puberty, followed by impaired bipotent TEC progenitor differentiation and depletion of Sca-1lo cTEC and mTEC lineage-specific precursors. We reveal that an increase in follistatin production by aging TECs contributes to their own demise. TEC loss occurs primarily through the antagonism of activin A signaling, which we show is required for TEC maturation and acts in dissonance to BMP4, which promotes the maintenance of TEC progenitors. These results support a model in which an imbalance of activin A and BMP4 signaling underpins the degeneration of postnatal TEC maintenance during aging, and its reversal enables the transient replenishment of mature TECs.


Activins/metabolism , Aging/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation , Epithelial Cells/metabolism , Follistatin/metabolism , Signal Transduction , Stem Cells/metabolism , Thymus Gland/metabolism , Animals , Epithelial Cells/cytology , Female , Mice , Mice, Inbred BALB C , Stem Cells/cytology , Thymus Gland/cytology
4.
Eur J Immunol ; 47(2): 291-304, 2017 02.
Article En | MEDLINE | ID: mdl-27861793

Although forkhead-box n1 (Foxn1) is a critical thymic epithelial cell regulator in thymus organogenesis, its association with epithelial differentiation and homeostasis in the postnatal and aged thymic microenvironment remains conflicting. Consequently, we have generated a Foxn1eGFP/+ knock-in mouse model that allows for refined investigation of the aging thymic epithelium. This reporter line differs from those previously published in that concomitant expression of enhanced green fluorescent protein enables live cell sorting of Foxn1+ cell populations. Our heterozygotes did not exhibit haploinsufficiency, with Foxn1 expression resembling that of wild-type mice. Comparative analysis between Foxn1 and enhanced green fluorescent protein at both the transcriptional and translational levels revealed co-localization, with progressive down-regulation observed predominantly in the aging cortical epithelium. Supplementation with bone morphogenetic protein (Bmp)-4 enhanced Foxn1 expression and colony forming efficiency in both embryonic and adult progenitor 3D cultures. Strikingly, selective maintenance of immature cortical and medullary epithelial cells was observed which is consistent with the higher Bmp receptor 2 expression levels seen in these progenitor populations. This study demonstrates the significance of our mouse model in unraveling the role of this master regulator in thymus development, homeostasis and aging, providing a faithful reporter system for phenotypic and functional investigations.


Aging/physiology , Epithelial Cells/physiology , Forkhead Transcription Factors/metabolism , Stem Cells/physiology , Thymus Gland/physiology , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cells, Cultured , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Homeostasis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Thymus Gland/cytology
5.
Stem Cell Reports ; 4(3): 445-58, 2015 Mar 10.
Article En | MEDLINE | ID: mdl-25733018

Mechanisms underlying age-related defects within lymphoid-lineages remain poorly understood. We previously reported that sex steroid ablation (SSA) induced lymphoid rejuvenation and enhanced recovery from hematopoietic stem cell (HSC) transplantation (HSCT). We herein show that, mechanistically, SSA induces hematopoietic and lymphoid recovery by functionally enhancing both HSC self-renewal and propensity for lymphoid differentiation through intrinsic molecular changes. Our transcriptome analysis revealed further hematopoietic support through rejuvenation of the bone marrow (BM) microenvironment, with upregulation of key hematopoietic factors and master regulatory factors associated with aging such as Foxo1. These studies provide important cellular and molecular insights into understanding how SSA-induced regeneration of the hematopoietic compartment can underpin recovery of the immune system following damaging cytoablative treatments. These findings support a short-term strategy for clinical use of SSA to enhance the production of lymphoid cells and HSC engraftment, leading to improved outcomes in adult patients undergoing HSCT and immune depletion in general.


Cell Differentiation , Gonadal Steroid Hormones/antagonists & inhibitors , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lymphopoiesis/physiology , Regeneration , Animals , Cell Count , Cell Differentiation/genetics , Cell Movement , Cell Self Renewal , Gene Expression Profiling , Gene Expression Regulation, Developmental , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Regeneration/genetics , Stem Cell Niche
6.
Cell Rep ; 8(4): 1198-209, 2014 Aug 21.
Article En | MEDLINE | ID: mdl-25131206

Thymic epithelial cells (TECs) are critical for T cell development and self-tolerance but are gradually lost with age. The existence of thymic epithelial progenitors (TEPCs) in the postnatal thymus has been inferred, but their identity has remained enigmatic. Here, we assessed the entire adult TEC compartment in order to reveal progenitor capacity is retained exclusively within a subset of immature thymic epithelium displaying several hallmark features of stem/progenitor function. These adult TEPCs generate mature cortical and medullary lineages in a stepwise fashion, including Aire+ TEC, within fetal thymus reaggregate grafts. Although relatively quiescent in vivo, adult TEPCs demonstrate significant in vitro colony formation and self-renewal. Importantly, 3D-cultured TEPCs retain their capacity to differentiate into cortical and medullary TEC lineages when returned to an in vivo thymic microenvironment. No other postnatal TEC subset exhibits this combination of properties. The characterization of adult TEPC will enable progress in understanding TEC biology in aging and regeneration.


Adult Stem Cells/physiology , Thymus Gland/cytology , Animals , Cell Lineage , Cell Proliferation , Cells, Cultured , Epithelial Cells/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic
7.
J Am Soc Nephrol ; 24(4): 573-85, 2013 Mar.
Article En | MEDLINE | ID: mdl-23393320

Loss of tolerance to neutrophil myeloperoxidase (MPO) underlies the development of ANCA-associated vasculitis and GN, but the mechanisms underlying this loss of tolerance are poorly understood. Here, we assessed the role of the thymus in deletion of autoreactive anti-MPO T cells and the importance of peripheral regulatory T cells in maintaining tolerance to MPO and protecting from GN. Thymic expression of MPO mRNA predominantly localized to medullary thymic epithelial cells. To assess the role of MPO in forming the T cell repertoire and the role of the autoimmune regulator Aire in thymic MPO expression, we compared the effects of immunizing Mpo(-/-) mice, Aire(-/-) mice, and control littermates with MPO. Immunized Mpo(-/-) and Aire(-/-) mice developed significantly more proinflammatory cytokine-producing anti-MPO T cells and higher ANCA titers than control mice. When we triggered GN with a subnephritogenic dose of anti-glomerular basement membrane antibody, Aire(-/-) mice had more severe renal disease than Aire(+/+) mice, consistent with a role for Aire-dependent central deletion in establishing tolerance to MPO. Furthermore, depleting peripheral regulatory T cells in wild-type mice also led to more anti-MPO T cells, higher ANCA titers, and more severe GN after immunization with MPO. Taken together, these results suggest that Aire-dependent central deletion and regulatory T cell-mediated peripheral tolerance both play major roles in establishing and maintaining tolerance to MPO, thereby protecting against the development of anti-MPO GN.


Antibodies, Antineutrophil Cytoplasmic/immunology , Glomerulonephritis/immunology , Immune Tolerance/immunology , Kidney Glomerulus/immunology , Peroxidase/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/metabolism , Animals , Antibodies, Antineutrophil Cytoplasmic/metabolism , Cytokines/metabolism , Glomerulonephritis/metabolism , Mice , Peroxidase/metabolism , RNA, Messenger , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/immunology
8.
Methods Mol Biol ; 945: 251-72, 2013.
Article En | MEDLINE | ID: mdl-23097111

The thymus organ is composed of a three-dimensional (3D) network of adjoining epithelium and stromal cells. Bone marrow-derived T cell precursors, upon entering the thymus, interact with and migrate through this cellular network as they differentiate and mature. An essential component of the stroma is the thymic epithelial cells (TEC), which play a vital role in T cell development and induction of self-tolerance for adaptive immunity. TEC can be isolated from the embryonic and adult thymus by a series of gentle enzymatic digestions and characterized into discrete subpopulations based on their expression of surface markers by flow cytometry. Enrichment of adult TEC can be achieved by depletion of hematopoietic cells, allowing sufficient numbers to be purified for subsequent functional and molecular analysis. Although monolayer cultures have been used to study TEC phenotype and T cell interaction, methods that mimic the 3D thymic microenvironment, such as fetal and reaggregate thymic organ cultures, are more accurate for the analysis of TEC function and support more complete T cell development. Herein, we describe methods for the efficient isolation and enrichment of TEC for downstream analyses as well as the reaggregation of embryonic progenitor epithelium to form a functional thymus graft under the kidney capsule.


Cell Culture Techniques/methods , Epithelial Cells/cytology , Flow Cytometry/methods , Thymus Gland/cytology , Animals , Cell Aggregation , DNA, Complementary/biosynthesis , Dissection , Epithelial Cells/metabolism , Female , Fetus/cytology , Kidney/cytology , Leukocyte Common Antigens/metabolism , Mice , Polymerase Chain Reaction , Pregnancy , RNA/genetics , RNA/isolation & purification , Stem Cells/cytology , Suspensions , Thymus Gland/embryology , Thymus Gland/transplantation
9.
Blood ; 121(1): 118-28, 2013 Jan 03.
Article En | MEDLINE | ID: mdl-23152546

The atypical chemokine receptor CCX-CKR regulates bioavailability of CCL19, CCL21, and CCL25, homeostatic chemokines that play crucial roles in thymic lymphopoiesis. Deletion of CCX-CKR results in accelerated experimental autoimmunity induced by immunization. Here we show that CCX-CKR deletion also increases incidence of a spontaneous Sjögren's syndrome-like pathology, characterized by lymphocytic infiltrates in salivary glands and liver of CCX-CKR(-/-) mice, suggestive of a defect in self-tolerance when CCX-CKR is deleted. This prompted detailed examination of the thymus in CCX-CKR(-/-) mice. Negatively selected mature SP cells were less abundant in CCX-CKR(-/-) thymi, yet expansion of both DP and immature SP cells was apparent. Deletion of CCX-CKR also profoundly reduced proportions of DN3 thymocyte precursors and caused DN2 cells to accumulate within the medulla. These effects are likely driven by alterations in thymic stroma as CCX-CKR(-/-) mice have fewer cTECs per thymocyte, and cTECs express the highest level of CCX-CKR in the thymus. A profound decrease in CCL25 within the thymic cortex was observed in CCX-CKR(-/-) thymi, likely accounting for their defects in thymocyte distribution and frequency. These findings identify a novel role for CCX-CKR in regulating cTEC biology, which promotes optimal thymocyte development and selection important for self-tolerant adaptive immunity.


Autoimmunity , Lymphopoiesis , Receptors, Chemokine/deficiency , Thymocytes/pathology , Thymus Gland/pathology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Chemokines/metabolism , Chemokines, CC/biosynthesis , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunophenotyping , Kidney/pathology , Liver/pathology , Lymphopoiesis/genetics , Male , Mice , Mice, Knockout , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Receptors, Chemokine/physiology , Self Tolerance/genetics , Self Tolerance/immunology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , Stem Cells/classification , Stem Cells/metabolism , Stem Cells/pathology , Submandibular Gland/pathology
10.
J Immunol Methods ; 385(1-2): 23-34, 2012 Nov 30.
Article En | MEDLINE | ID: mdl-22910002

The reproducible isolation and accurate characterization of thymic epithelial cell (TEC) subsets is of critical importance to the ongoing study of thymopoiesis and its functional decline with age. The study of adult TEC, however, is significantly hampered due to the severely low stromal to hematopoietic cell ratio. Non-biased digestion and enrichment protocols are thus essential to ensure optimal cell yield and accurate representation of stromal subsets, as close as possible to their in vivo representation. Current digestion protocols predominantly involve diverse, relatively impure enzymatic variants of crude collagenase and collagenase/dispase (col/disp) preparations, which have variable efficacy and are often suboptimal in their ability to mediate complete digestion of thymus tissue. To address these issues we compared traditional col/disp preparations with the latest panel of Liberase products that contain a blend of highly purified collagenase and neutral protease enzymes. Liberase enzymes revealed a more rapid, complete dissociation of thymus tissue; minimizing loss of viability and increasing recovery of thymic stromal cell (TSC) elements. In particular, the recovery and viability of TEC, notably the rare cortical subsets, were significantly enhanced with Liberase products containing medium to high levels of thermolysin. The improved stromal dissociation led to numerically increased TEC yield and total TEC RNA isolated from pooled digests of adult thymus. Furthermore, the increased recovery of TEC enhanced resolution and quantification of TEC subsets in both adult and aged mice, facilitating flow cytometric analysis on a per thymus basis. We further refined the adult TEC phenotype by correlating surface expression of known TEC markers, with expression of intracellular epithelial lineage markers, Keratin 5 and Keratin 8. The data reveal more extensive expression of K8 than previously recognized and indicates considerable heterogeneity still exists within currently defined adult TEC subsets.


Cell Separation/methods , Enzymes/metabolism , Epithelial Cells/cytology , Thymus Gland/cytology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Survival/immunology , Collagenases/metabolism , Endopeptidases/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Immunophenotyping , Keratin-5/metabolism , Keratin-8/metabolism , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results , Stromal Cells/cytology , Stromal Cells/immunology , Stromal Cells/metabolism , Thermolysin/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Time Factors
11.
J Immunol ; 184(11): 6014-24, 2010 Jun 01.
Article En | MEDLINE | ID: mdl-20483779

Cytotoxic antineoplastic therapy is used to treat malignant disease but results in long-term immunosuppression in postpubertal and adult individuals, leading to increased incidence and severity of opportunistic infections. We have previously shown that sex steroid ablation (SSA) reverses immunodeficiencies associated with age and hematopoietic stem cell transplantation in both autologous and allogeneic settings. In this study, we have assessed the effects of SSA by surgical castration on T cell recovery of young male mice following cyclophosphamide treatment as a model for the impact of chemotherapy. SSA increased thymic cellularity, involving all of the thymocyte subsets and early T lineage progenitors. It also induced early repair of damage to the thymic stromal microenvironment, which is crucial to the recovery of a fully functional T cell-based immune system. These functional changes in thymic stromal subsets included enhanced production of growth factors and chemokines important for thymopoiesis, which preceded increases in both thymocyte and stromal cellularity. These effects collectively translated to an increase in peripheral and splenic naive T cells. In conclusion, SSA enhances T cell recovery following cyclophosphamide treatment of mice, at the level of the thymocytes and their stromal niches. This provides a new approach to immune reconstitution following antineoplastic therapy.


Antineoplastic Agents/toxicity , Cyclophosphamide/toxicity , Gonadal Steroid Hormones/immunology , Orchiectomy , T-Lymphocytes/immunology , Animals , Cell Separation , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Steroids , T-Lymphocytes/drug effects , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/immunology
12.
J Immunol ; 183(2): 823-31, 2009 Jul 15.
Article En | MEDLINE | ID: mdl-19564346

Immunosuppressive drugs and cytotoxic chemotherapy agents are designed to kill or suppress autoreactive, alloaggressive, or hyperinflammatory T cells, or disseminated malignancies. However, they also cause severe immunological side effects ranging from interrupted thymopoiesis and general immunodeficiency to, paradoxically, autoimmunity. Consistent with the cross-talk between thymocytes and stromal cells, we now show that these common therapeutic agents have major effects on murine thymic epithelial cells (TEC), crucially required to rebuild immunity posttreatment. We show that the immunosuppressant cyclosporine A, which has been linked to a thymus-dependent autoimmune syndrome in some patients, causes extensive loss of autoimmune regulator (Aire(+)) tolerance-inducing MHC class II(high) medullary TEC (mTEC(high)). Post-cyclosporine A, Aire expression was restored within 7 days. Full recovery of the mTEC(high) subset occurred within 10 days and was linked to a decrease in a relatively resistant MHC class II(low) mTEC subset (mTEC(low)), consistent with a previously described precursor-product relationship. Cyclophosphamide and dexamethasone caused more extensive ablation of thymocytes and stromal cells but again severely depleted tolerance-inducing mTEC(high). Together, these data show that Aire(+) mTECs are highly sensitive to damage and that mTEC regeneration follows a conserved pattern regardless of the treatment regimen used.


Cyclophosphamide/pharmacology , Cyclosporine/pharmacology , Dexamethasone/pharmacology , Epithelial Cells/immunology , Immune Tolerance/drug effects , Thymus Gland/cytology , Animals , Anti-Inflammatory Agents/pharmacology , Autoimmunity/drug effects , Epithelial Cells/physiology , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred C57BL , Regeneration , Stromal Cells , Transcription Factors , AIRE Protein
13.
J Immunol ; 182(5): 2690-9, 2009 Mar 01.
Article En | MEDLINE | ID: mdl-19234163

The thymic stromal niche normally directs the production and export of a self-tolerant T cell repertoire. Many models of spontaneous autoimmunity, however, develop thymic architectural abnormalities before disease onset. Although this is suspected to affect central tolerance induction, creating an autoimmune predisposition, in-depth analysis of the microenvironment within these thymi is lacking, such that the mechanisms and likely direct effects on the T cell repertoire are unknown or speculative. Here we show that NZB mice, the first described model for systemic autoimmunity, demonstrate a complex thymic phenotype, including a lack of the autoimmune regulator (Aire), early defects in thymic epithelial cell (TEC) expansion, and evidence for altered NF-kappaB2 signaling. Analysis of medullary TEC revealed a numerical loss of the Aire-expressing MHC class II(high) (mTEC-high) subset as well reduced Aire protein and mRNA per cell. RelB expression was also reduced, while chemokines CCL19 and CCL21 were increased. Unexpectedly, the proportion of cortex and medulla in the NZB mice was normal from 36 wk, despite worsening architectural abnormalities. These data show that the NZB defect is more complex than previously appreciated, segregating into early numerical TEC deficiencies that correct with age, late degeneration of the niche architecture that does not affect TEC number, and a persistent reduction in Aire and RelB expression per cell acquired upon mTEC-high differentiation.


Autoimmune Diseases/immunology , Disease Models, Animal , Down-Regulation/immunology , NF-kappa B p52 Subunit/physiology , Signal Transduction/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factors/metabolism , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Disease Progression , Down-Regulation/genetics , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunophenotyping , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred NZB , NF-kappa B p52 Subunit/antagonists & inhibitors , Signal Transduction/genetics , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Thymus Gland/pathology , Transcription Factors/biosynthesis , Transcription Factors/physiology , Transcription, Genetic/immunology , AIRE Protein
14.
Semin Immunopathol ; 30(4): 457-77, 2008 Dec.
Article En | MEDLINE | ID: mdl-18982327

Poor immune recovery is characteristic of bone marrow transplantation and leads to high levels of morbidity and mortality. The primary underlying cause is a compromised thymic function, resulting from age-induced atrophy and further compounded by the damaging effects of cytoablative conditioning regimes on thymic epithelial cells (TEC). Several strategies have been proposed to enhance T cell reconstitution. Some, such as the use of single biological agents, are currently being tested in clinical trials. However, a more rational approach to immune restoration will be to leverage the evolving repertoire of new technologies. Specifically, the combined targeting of TEC, thymocytes and peripheral T cells, together with the bone marrow niches, promises a more strategic clinical therapeutic platform.


Hematopoietic Stem Cell Transplantation , Immunity, Cellular/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Transplantation Conditioning/adverse effects , Animals , Clinical Trials as Topic , Epithelial Cells/cytology , Humans , T-Lymphocytes/cytology , Thymus Gland/cytology
15.
J Immunol Methods ; 329(1-2): 56-66, 2008 Jan 01.
Article En | MEDLINE | ID: mdl-17988680

The microenvironment of the thymus consists of functionally discrete niches composed of distinct stromal cell subsets. Clinically relevant changes affecting T-cell differentiation occur within these niches with age and injury caused by irradiation and chemotherapy treatments. The study of thymic stromal cells has been hampered by the technical difficulty in isolating significant numbers of this important population. Here we present an improved protocol for enzymatic isolation of stromal cells that enables comparative flow cytometric analyses and their purification for downstream cellular or molecular analysis. Fractions analyzed throughout enzymatic digestion of the thymus revealed that various stromal subsets are isolated at characteristic intervals. This highlights the importance of pooling all cells isolated from the thymus for numerical and phenotypic analysis to avoid biased representation of subpopulations. We also describe refined magnetic bead separation techniques that yield almost pure preparations of CD45(-) stroma. Sorting of these suspensions using defined markers enabled purification of the major epithelial subsets, confirmed by keratin staining and PCR analysis. This three-step procedure represents a rapid, reproducible method for the unbiased purification of the stromal cells that direct thymic T-cell differentiation.


Cell Separation , Epithelial Cells , Flow Cytometry , Stromal Cells , Thymus Gland/cytology , Animals , Epithelial Cells/chemistry , Epithelial Cells/immunology , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Immunomagnetic Separation , Keratins/analysis , Leukocyte Common Antigens/analysis , Mice , Mice, Inbred C57BL , Phenotype , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/chemistry , Stromal Cells/immunology , Temperature , Thymus Gland/chemistry , Thymus Gland/immunology
16.
J Immunol ; 178(11): 7473-84, 2007 Jun 01.
Article En | MEDLINE | ID: mdl-17513799

Delayed immune reconstitution in adult recipients of allogeneic hemopoietic stem cell transplantations (HSCT) is related to age-induced thymic atrophy. Overcoming this paucity of T cell function is a major goal of clinical research but in the context of allogeneic transplants, any strategy must not exacerbate graft-vs-host disease (GVHD) yet ideally retain graft-vs-tumor (GVT) effects. We have shown sex steroid ablation reverses thymic atrophy and enhances T cell recovery in aged animals and in congenic bone marrow (BM) transplant but the latter does not have the complications of allogeneic T cell reactivity. We have examined whether sex steroid ablation promoted hemopoietic and T cell recovery following allogeneic HSCT and whether this benefit was negated by enhanced GVHD. BM and thymic cell numbers were significantly increased at 14 and 28 days after HSCT in castrated mice compared with sham-castrated controls. In the thymus, the numbers of donor-derived thymocytes and dendritic cells were significantly increased after HSCT and castration; donor-derived BM precursors and developing B cells were also significantly increased. Importantly, despite restoring T cell function, sex steroid inhibition did not exacerbate the development of GVHD or ameliorate GVT activity. Finally, IL-7 treatment in combination with castration had an additive effect on thymic cellularity following HSCT. These results indicate that sex steroid ablation can profoundly enhance thymic and hemopoietic recovery following allogeneic HSCT without increasing GVHD and maintaining GVT.


Hematopoietic Stem Cell Transplantation , Orchiectomy , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/transplantation , Bone Marrow Transplantation/immunology , Combined Modality Therapy , Dendritic Cells/immunology , Dendritic Cells/transplantation , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/immunology , Interleukin-7/administration & dosage , Interleukin-7/deficiency , Interleukin-7/genetics , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Spleen/immunology , Spleen/transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Thymus Gland/immunology , Thymus Gland/transplantation , Transplantation, Homologous/immunology
17.
J Immunol ; 175(4): 2741-53, 2005 Aug 15.
Article En | MEDLINE | ID: mdl-16081852

The thymus undergoes age-related atrophy, coincident with increased circulating sex steroids from puberty. The impact of thymic atrophy is most profound in clinical conditions that cause a severe loss in peripheral T cells with the ability to regenerate adequate numbers of naive CD4+ T cells indirectly correlating with patient age. The present study demonstrates that androgen ablation results in the complete regeneration of the aged male mouse thymus, restoration of peripheral T cell phenotype and function and enhanced thymus regeneration following bone marrow transplantation. Importantly, this technique is also applicable to humans, with analysis of elderly males undergoing sex steroid ablation therapy for prostatic carcinoma, demonstrating an increase in circulating T cell numbers, particularly naive (TREC+) T cells. Collectively these studies represent a fundamentally new approach to treating immunodeficiency states in humans.


Androgen Antagonists/administration & dosage , Gonadotropin-Releasing Hormone/agonists , Lymphocyte Activation/physiology , Regeneration/physiology , Thymus Gland/physiology , Aged , Aging/physiology , Animals , Antigens, Ly/biosynthesis , Apoptosis/physiology , Atrophy , Bone Marrow Transplantation , Castration , Cell Differentiation/physiology , Cell Proliferation , Humans , Immunophenotyping , Lymphocyte Activation/drug effects , Lymphopoiesis/drug effects , Lymphopoiesis/physiology , Male , Membrane Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Proto-Oncogene Proteins c-kit/biosynthesis , Regeneration/drug effects , Stromal Cells/cytology , Stromal Cells/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/physiology , Thymus Gland/anatomy & histology , Thymus Gland/drug effects , Thymus Gland/pathology
...