Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Immunity ; 46(3): 504-515, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329707

ABSTRACT

Maturation and migration to lymph nodes (LNs) constitutes a central paradigm in conventional dendritic cell (cDC) biology but remains poorly defined in humans. Using our organ donor tissue resource, we analyzed cDC subset distribution, maturation, and migration in mucosal tissues (lungs, intestines), associated lymph nodes (LNs), and other lymphoid sites from 78 individuals ranging from less than 1 year to 93 years of age. The distribution of cDC1 (CD141hiCD13hi) and cDC2 (Sirp-α+CD1c+) subsets was a function of tissue site and was conserved between donors. We identified cDC2 as the major mature (HLA-DRhi) subset in LNs with the highest frequency in lung-draining LNs. Mature cDC2 in mucosal-draining LNs expressed tissue-specific markers derived from the paired mucosal site, reflecting their tissue-migratory origin. These distribution and maturation patterns were largely maintained throughout life, with site-specific variations. Our findings provide evidence for localized DC tissue surveillance and reveal a lifelong division of labor between DC subsets, with cDC2 functioning as guardians of the mucosa.


Subject(s)
Dendritic Cells/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Infant , Male , Middle Aged , Young Adult
2.
Clin Immunol ; 240: 109048, 2022 07.
Article in English | MEDLINE | ID: mdl-35644520

ABSTRACT

Interactions between B cells and CD4+ T cells play a central role in the development of Type 1 Diabetes (T1D). Two helper cell subsets, follicular (Tfh) and peripheral (Tph) helper T cells, are increased in patients with T1D but their role in driving B cell autoimmunity is undefined. We used a personalized immune (PI) mouse model to generate human immune systems de novo from hematopoietic stem cells (HSCs) of patients with T1D or from healthy controls (HCs). Both groups developed Tfh and Tph-like cells, and those with T1D-derived immune systems demonstrated increased numbers of Tph-like and Tfh cells compared to HC-derived PI mice. T1D-derived immune systems included increased proportions of unconventional memory CD27-IgD- B cells and reduced proportions of naïve B cells compared to HC PI mice, resembling changes reported for patients with systemic lupus erythematosus. Our findings suggest that T1D HSCs are genetically programmed to produce increased proportions of T cells that promote the development of unconventional, possibly autoreactive memory B cells. PI mice provide an avenue for further understanding of the immune abnormalities that drive autoantibody pathogenesis and T1D.


Subject(s)
B-Lymphocyte Subsets , Diabetes Mellitus, Type 1 , Animals , Autoimmunity , B-Lymphocyte Subsets/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , T Follicular Helper Cells , T-Lymphocytes, Helper-Inducer
3.
Am J Transplant ; 21(4): 1465-1476, 2021 04.
Article in English | MEDLINE | ID: mdl-33021057

ABSTRACT

T cells are implicated in the pathogenesis of cardiac allograft vasculopathy (CAV), yet their clonality, specificity, and function are incompletely defined. Here we used T cell receptor ß chain (TCRB) sequencing to study the T cell repertoire in the coronary artery, endomyocardium, and peripheral blood at the time of retransplant in four cases of CAV and compared it to the immunoglobulin heavy chain variable region (IGHV) repertoire from the same samples. High-dimensional flow cytometry coupled with single-cell PCR was also used to define the T cell phenotype. Extensive overlap was observed between intragraft and blood TCRBs in all cases, a finding supported by robust quantitative diversity metrics. In contrast, blood and graft IGHV repertoires from the same samples showed minimal overlap. Coronary infiltrates included CD4+ and CD8+ memory T cells expressing inflammatory (IFNγ, TNFα) and profibrotic (TGFß) cytokines. These were distinguishable from the peripheral blood based on memory, activation, and tissue residency markers (CD45RO, CTLA-4, and CD69). Importantly, high-frequency rearrangements were traced back to endomyocardial biopsies (2-6 years prior). Comparison with four HLA-mismatched blood donors revealed a repertoire of shared TCRBs, including a subset of recently described cross-reactive sequences. These findings provide supportive evidence for an active local intragraft bystander T cell response in late-stage CAV.


Subject(s)
Heart Transplantation , Allografts , Coronary Vessels , Graft Rejection/etiology , Heart Transplantation/adverse effects , Humans , T-Lymphocytes
4.
Am J Transplant ; 20(1): 88-100, 2020 01.
Article in English | MEDLINE | ID: mdl-31319439

ABSTRACT

Siplizumab, a humanized anti-CD2 monoclonal antibody, has been used in conditioning regimens for hematopoietic cell transplantation and tolerance induction with combined kidney-bone marrow transplantation. Siplizumab-based tolerance induction regimens deplete T cells globally while enriching regulatory T cells (Tregs) early posttransplantation. Siplizumab inhibits allogeneic mixed-lymphocyte reactions (MLRs) in vitro. We compared the impact of siplizumab on Tregs versus other T cell subsets in HLA-mismatched allogeneic MLRs using PBMCs. Siplizumab predominantly reduced the percentage of CD4+ and CD8+ effector memory T cells, which express higher CD2 levels than naïve T cells or resting Tregs. Conversely, siplizumab enriched proliferating CD45RA- FoxP3HI cells in MLRs. FoxP3 expression was stable over time in siplizumab-containing cultures, consistent with enrichment for bona fide Tregs. Consistently, high-throughput TCRß CDR3 sequencing of sorted unstimulated and proliferating T cells in MLRs revealed selective expansion of donor-reactive Tregs along with depletion of donor-reactive CD4+ effector/memory T cells in siplizumab-containing MLRs. These results indicate that siplizumab may have immunomodulatory functions that may contribute to its success in tolerance-inducing regimens. Our studies also confirm that naïve in addition to effector/memory T cells contribute to the allogeneic MLR and mandate further investigation of the impact of siplizumab on alloreactive naïve T cells.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Immune Tolerance/immunology , Immunologic Memory/immunology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Humans , Immune Tolerance/drug effects , Immunologic Memory/drug effects , In Vitro Techniques , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects
5.
J Immunol ; 201(2): 432-439, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29848753

ABSTRACT

Viral respiratory tract infections (VRTI) remain a leading cause of morbidity and mortality among infants and young children. In mice, optimal protection to VRTI is mediated by recruitment of effector T cells to the lungs and respiratory tract, and subsequent establishment of tissue resident memory T cells (Trm), which provide long-term protection. These critical processes of T cell recruitment to the respiratory tract, their role in disease pathogenesis, and establishment of local protective immunity remain undefined in pediatric VRTI. In this study, we investigated T cell responses in the upper respiratory tract (URT) and lower respiratory tract (LRT) of infants and young children with VRTI, revealing developmental regulation of T cell differentiation and Trm generation in situ. We show a direct concurrence between T cell responses in the URT and LRT, including a preponderance of effector CD8+ T cells that was associated with disease severity. During infant VRTI, there was an accumulation of terminally differentiated effector cells (effector memory RA+ T cells) in the URT and LRT with reduced Trm in the early neonatal period, and decreased effector memory RA+ T cell and increased Trm formation with age during the early years of childhood. Moreover, human infant T cells exhibit increased expression of the transcription factor T-bet compared with adult T cells, suggesting a mechanism for preferential generation of effector over Trm. The developmental regulation of respiratory T cell responses as revealed in the present study is important for diagnosing, monitoring, and treating VRTI in the critical early life stages.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Respiratory Tract Infections/immunology , Virus Diseases/immunology , Adult , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Lung/immunology , Lung/virology , Lymphocyte Activation/immunology , Male , Respiratory Tract Infections/virology , Transcription Factors/immunology
6.
Am J Transplant ; 19(8): 2186-2198, 2019 08.
Article in English | MEDLINE | ID: mdl-30768842

ABSTRACT

Reliable in vitro expansion protocols of regulatory T cells (Tregs) are needed for clinical use. We studied the biology of Mauritian Cynomolgus macaque (MCM) Tregs and developed four in vitro Treg expansion protocols for translational studies. Tregs expanded 3000-fold when artificial antigen presenting cells (aAPCs) expressing human CD80, CD58 and CD32 were used throughout the culture. When donor peripheral blood mononuclear cells (PBMCs) were used as the single source of APCs followed by aAPCs, Tregs expanded 2000-fold. Tregs from all protocols suppressed the proliferation of anti-CD2CD3CD28 bead-stimulated autologous PBMCs albeit with different potencies, varying from 1:2-1:4 Treg:PBMC ratios, up to >1:32. Reculture of cryopreserved Tregs permitted reexpansion with improved suppressive activity. Occasionally, CD8 contamination was observed and resolved by resorting. Specificity studies showed greater suppression of stimulation by anti-CD2CD3CD28 beads of PBMCs from the same donor used for stimulation during the Treg cultures and of autologous cells than of third-party PBMC responders. Similar to humans, the Treg-specific demethylated region (TSDR) within the Foxp3 locus correlated with suppressive activity and expression of Foxp3. Contrary to humans, FoxP3 expression did not correlate with CD45RA or CD127 expression. In summary, we have characterized MCM Tregs and developed four Treg expansion protocols that can be used for preclinical applications.


Subject(s)
Antigen-Presenting Cells/immunology , DNA Methylation , Forkhead Transcription Factors/metabolism , Leukocytes, Mononuclear/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Forkhead Transcription Factors/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macaca fascicularis , T-Lymphocytes, Regulatory/metabolism
7.
Xenotransplantation ; 25(5): e12391, 2018 09.
Article in English | MEDLINE | ID: mdl-29527745

ABSTRACT

BACKGROUND: Despite recent progress in survival times of xenografts in non-human primates, there are no reports of survival beyond 5 days of histologically well-aerated porcine lung grafts in baboons. Here, we report our initial results of pig-to-baboon xeno-lung transplantation (XLTx). METHODS: Eleven baboons received genetically modified porcine left lungs from either GalT-KO alone (n = 3), GalT-KO/humanCD47(hCD47)/hCD55 (n = 3), GalT-KO/hD47/hCD46 (n = 4), or GalT-KO/hCD39/hCD46/hCD55/TBM/EPCR (n = 1) swine. The first 2 XLTx procedures were performed under a non-survival protocol that allowed a 72-hour follow-up of the recipients with general anesthesia, while the remaining 9 underwent a survival protocol with the intention of weaning from ventilation. RESULTS: Lung graft survivals in the 2 non-survival animals were 48 and >72 hours, while survivals in the other 9 were 25 and 28 hours, at 5, 5, 6, 7, >7, 9, and 10 days. One baboon with graft survival >7 days, whose entire lung graft remained well aerated, was euthanized on POD 7 due to malfunction of femoral catheters. hCD47 expression of donor lungs was detected in both alveoli and vessels only in the 3 grafts surviving >7, 9, and 10 days. All other grafts lacked hCD47 expression in endothelial cells and were completely rejected with diffuse hemorrhagic changes and antibody/complement deposition detected in association with early graft loss. CONCLUSIONS: To our knowledge, this is the first evidence of histologically viable porcine lung grafts beyond 7 days in baboons. Our results indicate that GalT-KO pig lungs are highly susceptible to acute humoral rejection and that this may be mitigated by transgenic expression of hCD47.


Subject(s)
Animals, Genetically Modified/immunology , CD47 Antigen/immunology , Graft Rejection/immunology , Graft Survival/immunology , Papio/immunology , Animals , Graft Rejection/pathology , Heterografts/immunology , Humans , Lung/immunology , Lung Transplantation/methods , Swine , Transplantation, Heterologous/methods , Transplants/immunology
8.
Nat Commun ; 12(1): 5761, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599177

ABSTRACT

The thymus is a central lymphoid organ primarily responsible for the development of T cells. A small proportion of B cells, however, also reside in the thymus to assist negative selection of self-reactive T cells. Here we show that the thymus of human neonates contains a consistent contingent of CD138+ plasma cells, producing all classes and subclasses of immunoglobulins with the exception of IgD. These antibody-secreting cells are part of a larger subset of B cells that share the expression of signature genes defining mouse B1 cells, yet lack the expression of complement receptors CD21 and CD35. Data from single-cell transcriptomic, clonal correspondence and in vitro differentiation assays support the notion of intrathymic CD138+ plasma cell differentiation, alongside other B cell subsets with distinctive molecular phenotypes. Lastly, neonatal thymic plasma cells also include clones reactive to commensal and pathogenic bacteria that commonly infect children born with antibody deficiency. Thus, our findings point to the thymus as a source of innate humoral immunity in human neonates.


Subject(s)
Cell Differentiation , Plasma Cells/cytology , Thymus Gland/cytology , Adult , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/ultrastructure , Fetal Blood/cytology , Gene Expression Profiling , Humans , Immunity, Innate , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Infant, Newborn , Lymphocyte Subsets/cytology , Principal Component Analysis , RNA-Seq , Single-Cell Analysis , Somatic Hypermutation, Immunoglobulin/genetics , Transcriptome/genetics
9.
Elife ; 102021 10 26.
Article in English | MEDLINE | ID: mdl-34698636

ABSTRACT

Using a high-throughput mitochondrial phenotyping platform to quantify multiple mitochondrial features among molecularly defined immune cell subtypes, we quantify the natural variation in mitochondrial DNA copy number (mtDNAcn), citrate synthase, and respiratory chain enzymatic activities in human neutrophils, monocytes, B cells, and naïve and memory T lymphocyte subtypes. In mixed peripheral blood mononuclear cells (PBMCs) from the same individuals, we show to what extent mitochondrial measures are confounded by both cell type distributions and contaminating platelets. Cell subtype-specific measures among women and men spanning four decades of life indicate potential age- and sex-related differences, including an age-related elevation in mtDNAcn, which are masked or blunted in mixed PBMCs. Finally, a proof-of-concept, repeated-measures study in a single individual validates cell type differences and also reveals week-to-week changes in mitochondrial activities. Larger studies are required to validate and mechanistically extend these findings. These mitochondrial phenotyping data build upon established immunometabolic differences among leukocyte subpopulations, and provide foundational quantitative knowledge to develop interpretable blood-based assays of mitochondrial health.


Subject(s)
Leukocytes, Mononuclear/physiology , Memory T Cells/immunology , Mitochondria/metabolism , Monocytes/immunology , Neutrophils/immunology , Phenotype , Adult , Age Factors , Female , Humans , Male , Middle Aged , Sex Factors , Young Adult
10.
J Clin Invest ; 131(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33630757

ABSTRACT

In humans receiving intestinal transplantation (ITx), long-term multilineage blood chimerism often develops. Donor T cell macrochimerism (≥4%) frequently occurs without graft-versus-host disease (GVHD) and is associated with reduced rejection. Here we demonstrate that patients with macrochimerism had high graft-versus-host (GvH) to host-versus-graft (HvG) T cell clonal ratios in their allografts. These GvH clones entered the circulation, where their peak levels were associated with declines in HvG clones early after transplant, suggesting that GvH reactions may contribute to chimerism and control HvG responses without causing GVHD. Consistently, donor-derived T cells, including GvH clones, and CD34+ hematopoietic stem and progenitor cells (HSPCs) were simultaneously detected in the recipients' BM more than 100 days after transplant. Individual GvH clones appeared in ileal mucosa or PBMCs before detection in recipient BM, consistent with an intestinal mucosal origin, where donor GvH-reactive T cells expanded early upon entry of recipient APCs into the graft. These results, combined with cytotoxic single-cell transcriptional profiles of donor T cells in recipient BM, suggest that tissue-resident GvH-reactive donor T cells migrated into the recipient circulation and BM, where they destroyed recipient hematopoietic cells through cytolytic effector functions and promoted engraftment of graft-derived HSPCs that maintain chimerism. These mechanisms suggest an approach to achieving intestinal allograft tolerance.


Subject(s)
Graft vs Host Disease/immunology , Intestines/transplantation , Lymphopoiesis/immunology , Organ Transplantation , T-Lymphocytes/immunology , Transplantation Chimera/immunology , Allografts , Female , Graft vs Host Disease/pathology , Humans , Intestines/immunology , Intestines/pathology , Male , T-Lymphocytes/pathology
11.
J Virol ; 82(14): 7089-99, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18480460

ABSTRACT

To examine the pathway of the coreceptor switching of CCR5-using (R5) virus to CXCR4-using (X4) virus in simian-human immunodeficiency virus SHIV(SF162P3N)-infected rhesus macaque BR24, analysis was performed on variants present at 20 weeks postinfection, the time when the signature gp120 V3 loop sequence of the X4 switch variant was first detected by PCR. Unexpectedly, circulating and tissue variants with His/Ile instead of the signature X4 V3 His/Arg insertions predominated at this time point. Phylogenetic analysis of the sequences of the C2 conserved region to the V5 variable loop of the envelope (Env) protein showed that viruses bearing HI insertions represented evolutionary intermediates between the parental SHIV(SF162P3N) and the final X4 HR switch variant. Functional analyses demonstrated that the HI variants were phenotypic intermediates as well, capable of using both CCR5 and CXCR4 for entry. However, the R5X4 intermediate virus entered CCR5-expressing target cells less efficiently than the parental R5 strain and was more sensitive to both CCR5 and CXCR4 inhibitors than either the parental R5 or the final X4 virus. It was also more sensitive than the parental R5 virus to antibody neutralization, especially to agents directed against the CD4 binding site, but not as sensitive as the late X4 virus. Significantly, the V3 loop sequence that determined CXCR4 use also conferred soluble CD4 neutralization sensitivity. Collectively, the data illustrate that, similar to human immunodeficiency virus type 1 (HIV-1) infection in individuals, the evolution from CCR5 to CXCR4 usage in BR24 transitions through an intermediate phase with reduced virus entry and coreceptor usage efficiencies. The data further support a model linking an open envelope gp120 conformation, better CD4 binding, and expansion to CXCR4 usage.


Subject(s)
Membrane Glycoproteins/genetics , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Viral Envelope Proteins/genetics , Amino Acid Substitution/genetics , Animals , Anti-HIV Agents/pharmacology , Humans , Macaca mulatta , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mutation, Missense , Neutralization Tests , Phylogeny , Polymerase Chain Reaction , RNA, Viral/genetics , Sequence Analysis, DNA , Sequence Homology , Simian Acquired Immunodeficiency Syndrome/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Virus Internalization
12.
J Virol ; 82(11): 5653-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18385246

ABSTRACT

We report here a second case of coreceptor switch in R5 simian-human immunodeficiency virus SF162P3N (SHIV(SF162P3N))-infected macaque CA28, supporting the use of this experimental system to examine factors that drive the change in coreceptor preference in vivo. Virus recovered from CA28 plasma (SHIV(CA28NP)) used both CCR5 and CXCR4 for entry, but the virus recovered from lymph node (SHIV(CA28NL)) used CXCR4 almost exclusively. Sequence and functional analyses showed that mutations in the V3 loop that conferred CXCR4 usage in macaque CA28 differed from those described in the previously reported case, demonstrating divergent mutational pathways for change in the coreceptor preference of the R5 SHIV(SF162P3N) isolate in vivo.


Subject(s)
Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Count , Macaca mulatta/genetics , Macaca mulatta/metabolism , Macaca mulatta/virology , Molecular Sequence Data , Mutation/genetics , Simian Immunodeficiency Virus/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
13.
J Clin Invest ; 129(6): 2446-2462, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30920391

ABSTRACT

We investigated human T-cell repertoire formation using high throughput TCRß CDR3 sequencing in immunodeficient mice receiving human hematopoietic stem cells (HSCs) and human thymus grafts. Replicate humanized mice generated diverse and highly divergent repertoires. Repertoire narrowing and increased CDR3ß sharing was observed during thymocyte selection. While hydrophobicity analysis implicated self-peptides in positive selection of the overall repertoire, positive selection favored shorter shared sequences that had reduced hydrophobicity at positions 6 and 7 of CDR3ßs, suggesting weaker interactions with self-peptides than unshared sequences, possibly allowing escape from negative selection. Sharing was similar between autologous and allogeneic thymi and occurred between different cell subsets. Shared sequences were enriched for allo-crossreactive CDR3ßs and for Type 1 diabetes-associated autoreactive CDR3ßs. Single-cell TCR-sequencing showed increased sharing of CDR3αs compared to CDR3ßs between mice. Our data collectively implicate preferential positive selection for shared human CDR3ßs that are highly cross-reactive. While previous studies suggested a role for recombination bias in producing "public" sequences in mice, our study is the first to demonstrate a role for thymic selection. Our results implicate positive selection for promiscuous TCRß sequences that likely evade negative selection, due to their low affinity for self-ligands, in the abundance of "public" human TCRß sequences.


Subject(s)
Complementarity Determining Regions , Receptors, Antigen, T-Cell, alpha-beta , Thymocytes/immunology , Thymus Gland/immunology , Animals , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Cross Reactions , Humans , Mice , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Thymocytes/cytology , Thymus Gland/cytology
14.
Cell Stem Cell ; 24(2): 227-239.e8, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30503142

ABSTRACT

Human intestinal transplantation often results in long-term mixed chimerism of donor and recipient blood in transplant patients. We followed the phenotypes of chimeric peripheral blood cells in 21 patients receiving intestinal allografts over 5 years. Donor lymphocyte phenotypes suggested a contribution of hematopoietic stem and progenitor cells (HSPCs) from the graft. Surprisingly, we detected donor-derived HSPCs in intestinal mucosa, Peyer's patches, mesenteric lymph nodes, and liver. Human gut HSPCs are phenotypically similar to bone marrow HSPCs and have multilineage differentiation potential in vitro and in vivo. Analysis of circulating post-transplant donor T cells suggests that they undergo selection in recipient lymphoid organs to acquire immune tolerance. Our longitudinal study of human HSPCs carried in intestinal allografts demonstrates their turnover kinetics and gradual replacement of donor-derived HSPCs from a circulating pool. Thus, we have demonstrated the existence of functioning HSPCs in human intestines with implications for promoting tolerance in transplant recipients.


Subject(s)
Cell Movement , Hematopoietic Stem Cells/cytology , Intestines/cytology , Intestines/transplantation , Animals , Cell Differentiation , Cell Line , Cell Lineage , Chimerism , Graft vs Host Disease/immunology , Humans , Immune Tolerance , Intestinal Mucosa/cytology , Liver/cytology , Lymph Nodes/cytology , Mice , Peyer's Patches/cytology , Phenotype , T-Lymphocytes/cytology , Tissue Donors , Transplantation, Homologous
15.
JCI Insight ; 3(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30429370

ABSTRACT

Allograft tolerance, in which a graft is accepted without long-term immunosuppression, could overcome numerous obstacles in transplantation. Human allograft tolerance has been intentionally induced across HLA barriers via combined kidney and bone marrow transplantation (CKBMT) with a regimen that induces only transient chimerism. Tregs are enriched early after CKBMT. While deletional tolerance contributes to long-term tolerance, the role of Tregs remains unclear. We have optimized a method for identifying the donor-specific Treg repertoire and used it to interrogate the fate of donor-specific Tregs after CKBMT. We expanded Tregs with several different protocols. Using functional analyses and T cell receptor sequencing, we found that expanding sorted Tregs with activated donor B cells identified the broadest Treg repertoire with the greatest potency and donor specificity of suppression. This method outperformed both alloantigen stimulation with CTLA4Ig and sequencing of CFSElo cells from the primary mixed lymphocyte reaction. In 3 tolerant and 1 nontolerant CKBMT recipients, we sequenced donor-specific Tregs before transplant and tracked them after transplant. Preexisting donor-specific Tregs were expanded at 6 months after CKBMT in tolerant patients and were reduced in the nontolerant patient. These results suggest that early expansion of donor-specific Tregs is involved in tolerance induction following CKBMT.


Subject(s)
Kidney Transplantation , T-Lymphocytes, Regulatory/transplantation , Transplantation Tolerance , B-Lymphocytes/immunology , B-Lymphocytes/transplantation , Bone Marrow Transplantation , CD4 Lymphocyte Count , CTLA-4 Antigen/immunology , Humans , Lymphocyte Culture Test, Mixed , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , Tissue Donors
16.
Cell Rep ; 20(12): 2921-2934, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930685

ABSTRACT

Tissue-resident memory T cells (TRMs) in mice mediate optimal protective immunity to infection and vaccination, while in humans, the existence and properties of TRMs remain unclear. Here, we use a unique human tissue resource to determine whether human tissue memory T cells constitute a distinct subset in diverse mucosal and lymphoid tissues. We identify a core transcriptional profile within the CD69+ subset of memory CD4+ and CD8+ T cells in lung and spleen that is distinct from that of CD69- TEM cells in tissues and circulation and defines human TRMs based on homology to the transcriptional profile of mouse CD8+ TRMs. Human TRMs in diverse sites exhibit increased expression of adhesion and inhibitory molecules, produce both pro-inflammatory and regulatory cytokines, and have reduced turnover compared with circulating TEM, suggesting unique adaptations for in situ immunity. Together, our results provide a unifying signature for human TRM and a blueprint for designing tissue-targeted immunotherapies.


Subject(s)
Gene Expression Profiling , Immunologic Memory , Lymphoid Tissue/immunology , Mucous Membrane/immunology , T-Lymphocytes/immunology , Transcription, Genetic , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Lineage/genetics , Clone Cells , Humans , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Mice , Phenotype , Transcriptome/genetics
17.
Nat Cell Biol ; 19(5): 542-549, 2017 May.
Article in English | MEDLINE | ID: mdl-28436965

ABSTRACT

Recapitulation of lung development from human pluripotent stem cells (hPSCs) in three dimensions (3D) would allow deeper insight into human development, as well as the development of innovative strategies for disease modelling, drug discovery and regenerative medicine. We report here the generation from hPSCs of lung bud organoids (LBOs) that contain mesoderm and pulmonary endoderm and develop into branching airway and early alveolar structures after xenotransplantation and in Matrigel 3D culture. Expression analysis and structural features indicated that the branching structures reached the second trimester of human gestation. Infection in vitro with respiratory syncytial virus, which causes small airway obstruction and bronchiolitis in infants, led to swelling, detachment and shedding of infected cells into the organoid lumens, similar to what has been observed in human lungs. Introduction of mutation in HPS1, which causes an early-onset form of intractable pulmonary fibrosis, led to accumulation of extracellular matrix and mesenchymal cells, suggesting the potential use of this model to recapitulate fibrotic lung disease in vitro. LBOs therefore recapitulate lung development and may provide a useful tool to model lung disease.


Subject(s)
Cell Culture Techniques , Lung/pathology , Organoids/pathology , Pluripotent Stem Cells/pathology , Pulmonary Fibrosis/pathology , Respiratory Syncytial Virus Infections/pathology , Tissue Engineering/methods , Animals , Cell Differentiation , Cells, Cultured , Female , Genetic Predisposition to Disease , Humans , Lung/metabolism , Lung/virology , Lung Transplantation , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred NOD , Mutation , Organogenesis , Organoids/metabolism , Organoids/transplantation , Organoids/virology , Phenotype , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , Pluripotent Stem Cells/virology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Time Factors
18.
Cell Stem Cell ; 21(6): 747-760.e7, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29198940

ABSTRACT

Myeloid-biased hematopoietic stem cells (MB-HSCs) play critical roles in recovery from injury, but little is known about how they are regulated within the bone marrow niche. Here we describe an auto-/paracrine physiologic circuit that controls quiescence of MB-HSCs and hematopoietic progenitors marked by histidine decarboxylase (Hdc). Committed Hdc+ myeloid cells lie in close anatomical proximity to MB-HSCs and produce histamine, which activates the H2 receptor on MB-HSCs to promote their quiescence and self-renewal. Depleting histamine-producing cells enforces cell cycle entry, induces loss of serial transplant capacity, and sensitizes animals to chemotherapeutic injury. Increasing demand for myeloid cells via lipopolysaccharide (LPS) treatment specifically recruits MB-HSCs and progenitors into the cell cycle; cycling MB-HSCs fail to revert into quiescence in the absence of histamine feedback, leading to their depletion, while an H2 agonist protects MB-HSCs from depletion after sepsis. Thus, histamine couples lineage-specific physiological demands to intrinsically primed MB-HSCs to enforce homeostasis.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Histamine/metabolism , Myeloid Cells/metabolism , Animals , Bone Marrow/drug effects , Bone Marrow Transplantation , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Lipopolysaccharides/pharmacology , Mice , Myeloid Cells/drug effects
19.
Curr HIV Res ; 7(1): 30-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19149552

ABSTRACT

The human immunodeficiency virus (HIV) enters target cells via interaction of the viral glycoprotein with the cellular receptor CD4 and two principal coreceptors, CCR5 (R5 viruses) and CXCR4 (X4 viruses). Most HIV-1 transmissions result in a predominantly R5 virus infection. With time, X4 variants arise and coexist with R5 virus variants in approximately 50% of subtype B infected individuals. The underlying basis for virus coreceptor switch late in infection remains an enigma, but will be important to understand given that the appearance of X4 virus in HIV-1 infected patients inevitably heralds an unfavorable clinical outcome. Recently, emergence of X4 viruses was observed in rhesus macaques experimentally infected with a CCR5-tropic simian-human immunodeficiency virus (SHIV) with progression to disease, providing some insights into the process of coreceptor switching in vivo. Further studies in this animal model should enhance our understanding of the mechanistic basis for, and obstacles to, coreceptor switch.


Subject(s)
HIV-1/physiology , Receptors, HIV/metabolism , Simian Immunodeficiency Virus/physiology , Virus Attachment , Virus Internalization , Animals , HIV-1/genetics , Humans , Macaca mulatta , Simian Immunodeficiency Virus/genetics
20.
J Virol ; 81(16): 8621-33, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17537860

ABSTRACT

The basis for the switch from CCR5 to CXCR4 coreceptor usage seen in approximately 50% of human immunodeficiency virus type 1 (HIV-1) subtype B-infected individuals as disease advances is not well understood. Among the reasons proposed are target cell limitation and better immune recognition of the CXCR4 (X4)-tropic compared to the CCR5 (R5)-tropic virus. We document here X4 virus emergence in a rhesus macaque (RM) infected with R5-tropic simian/human immunodeficiency virus, demonstrating that coreceptor switch can happen in a nonhuman primate model of HIV/AIDS. The switch to CXCR4 usage in RM requires envelope sequence changes in the V3 loop that are similar to those found in humans, suggesting that the R5-to-X4 evolution pathways in the two hosts overlap. Interestingly, compared to the inoculating R5 virus, the emerging CXCR4-using virus is highly neutralization sensitive. This finding, coupled with the observation of X4 evolution and appearance in an animal with undetectable circulating virus-specific antibody and low cellular immune responses, lends further support to an inhibitory role of antiviral immunity in HIV-1 coreceptor switch.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Evolution, Molecular , HIV Infections/virology , HIV/genetics , Receptors, CCR5/physiology , Receptors, CXCR4/physiology , Simian Immunodeficiency Virus/genetics , Viral Envelope Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Viral/blood , Disease Models, Animal , HIV/isolation & purification , HIV/physiology , Macaca mulatta , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Phylogeny , Protein Structure, Secondary , Simian Immunodeficiency Virus/isolation & purification , Simian Immunodeficiency Virus/physiology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL