Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
1.
Cell Rep ; 32(2): 107892, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32668241

ABSTRACT

Tim-1, a phosphatidylserine receptor expressed on B cells, induces interleukin 10 (IL-10) production by sensing apoptotic cells. Here we show that mice with B cell-specific Tim-1 deletion develop tissue inflammation in multiple organs including spontaneous paralysis with inflammation in the central nervous system (CNS). Transcriptomic analysis demonstrates that besides IL-10, Tim-1+ B cells also differentially express a number of co-inhibitory checkpoint receptors including TIGIT. Mice with B cell-specific TIGIT deletion develop spontaneous paralysis with CNS inflammation, but with limited inflammation in other organs. Our findings suggest that Tim-1+ B cells are essential for maintaining self-tolerance and restraining tissue inflammation, and that Tim-1 signaling-dependent TIGIT expression on B cells is essential for maintaining CNS-specific tolerance. A possible critical role of aryl hydrocarbon receptor (AhR) in regulating the B cell function is discussed, as we find that AhR is among the preferentially expressed transcription factors in Tim-1+ B cells and regulates their TIGIT and IL-10 expression.


Subject(s)
B-Lymphocytes/metabolism , Hepatitis A Virus Cellular Receptor 1/metabolism , Inflammation/pathology , Organ Specificity , Receptors, Immunologic/metabolism , Aging/pathology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunomodulation , Interleukin-10/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments
2.
EBioMedicine ; 41: 571-583, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30833191

ABSTRACT

BACKGROUND: Acute T-cell mediated rejection (TCMR) is usually indicated by alteration in serum-creatinine measurements when considerable transplant damage has already occurred. There is, therefore, a need for non-invasive early detection of immune signals that would precede the onset of rejection, prior to transplant damage. METHODS: We examined the RT-qPCR expression of 22 literature-based genes in peripheral blood samples from 248 patients in the Kidney Allograft Immune Biomarkers of Rejection Episodes (KALIBRE) study. To account for post-transplantation changes unrelated to rejection, we generated time-adjusted gene-expression residuals from linear mixed-effects models in stable patients. To select genes, we used penalised logistic regression based on 27 stable patients and 27 rejectors with biopsy-proven T-cell-mediated rejection, fulfilling strict inclusion/exclusion criteria. We validated this signature in i) an independent group of stable patients and patients with concomitant T-cell and antibody-mediated-rejection, ii) patients from an independent study, iii) cross-sectional pre-biopsy samples from non-rejectors and iv) longitudinal follow-up samples covering the first post-transplant year from rejectors, non-rejectors and stable patients. FINDINGS: A parsimonious TCMR-signature (IFNG, IP-10, ITGA4, MARCH8, RORc, SEMA7A, WDR40A) showed cross-validated area-under-ROC curve 0.84 (0.77-0.88) (median, 2.5th-97.5th centile of fifty cross-validation cycles), sensitivity 0.67 (0.59-0.74) and specificity 0.85 (0.75-0.89). The estimated probability of TCMR increased seven weeks prior to the diagnostic biopsy and decreased after treatment. Gene expression in all patients showed pronounced variability, with up to 24% of the longitudinal samples in stable patients being TCMR-signature positive. In patients with borderline changes, up to 40% of pre-biopsy samples were TCMR-signature positive. INTERPRETATION: Molecular marker alterations in blood emerge well ahead of the time of clinically overt TCMR. Monitoring a TCMR-signature in peripheral blood could unravel T-cell-related pro-inflammatory activity and hidden immunological processes. This additional information could support clinical management decisions in cases of patients with stable but poor kidney function or with inconclusive biopsy results.


Subject(s)
Graft Rejection/etiology , Kidney Transplantation , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Antigens, CD/genetics , Area Under Curve , Cross-Sectional Studies , Female , GPI-Linked Proteins/genetics , Humans , Interferon-gamma/genetics , Kidney Transplantation/adverse effects , Longitudinal Studies , Male , Middle Aged , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Polyomavirus/pathogenicity , ROC Curve , Semaphorins/genetics , T-Lymphocytes/metabolism , Transcriptome , Young Adult
3.
Front Immunol ; 9: 2007, 2018.
Article in English | MEDLINE | ID: mdl-30233592

ABSTRACT

Hematopoietic stem/progenitor cells (HSPC) are characterized by their unique capacities of self-renewal and multi-differentiation potential. This second property makes them able to adapt their differentiation profile depending on the local environment they reach. Taking advantage of an animal model of peritonitis, induced by injection of the TLR-2 ligand, zymosan, we sought to study the relationship between bone marrow-derived hematopoietic stem/progenitor cells (BM-HSPCs) and innate lymphoid cells (ILCs) regarding their emergence and differentiation at the site of inflammation. Our results demonstrate that the strength of the inflammatory signals affects the capacity of BM-derived HSPCs to migrate and give rise in situ to ILCs. Both low- and high-dose of zymosan injections trigger the appearance of mature ILCs in the peritoneal cavity where the inflammation occurs. Herein, we show that only in low-dose injected mice, the recovered ILCs are dependent on an in situ differentiation of BM-derived HSPCs and/or ILC2 precursors (ILC2P) wherein high-dose, the stronger inflammatory environment seems to be able to induce the emergence of ILCs independently of BM-derived HSPCs. We suggest that a relationship between HSPCs and ILCs seems to be affected by the strength of the inflammatory stimuli opening new perspectives in the manipulation of these early hematopoietic cells.


Subject(s)
Hematopoietic Stem Cells/immunology , Lymphocytes/immunology , Peritonitis/immunology , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Movement , Cell Self Renewal , Cells, Cultured , Disease Models, Animal , Female , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Signal Transduction , Stem Cell Niche , Zymosan
4.
Pediatr Diabetes ; 19(5): 945-954, 2018 08.
Article in English | MEDLINE | ID: mdl-29473705

ABSTRACT

OBJECTIVE: To determine the safety and pharmacokinetics of alpha-1 antitrypsin (AAT) in adults and children. RESEARCH DESIGN AND METHODS: Short-term AAT treatment restores euglycemia in the non-obese mouse model of type 1 diabetes. A phase I multicenter study in 16 subjects with new-onset type 1 diabetes studied the safety and pharmacokinetics of Aralast NP (AAT). This open-label, dose-escalation study enrolled 8 adults aged 16 to 35 years and 8 children aged 8 to 15 years within 100 days of diagnosis, to receive 12 infusions of AAT: a low dose of 45 mg/kg weekly for 6 weeks, followed by a higher dose of 90 mg/kg for 6 weeks. RESULTS: C-peptide secretion during a mixed meal, hemoglobin A1c (HbA1c), and insulin usage remained relatively stable during the treatment period. At 72 hours after infusion of 90 mg/kg, mean levels of AAT fell below 2.0 g/L for 7 of 15 subjects. To identify a plasma level of AAT likely to be therapeutic, pharmacodynamic ex vivo assays were performed on fresh whole blood from adult subjects. Polymerase chain reaction (PCR) analyses were performed on inhibitor of IKBKE, NOD1, TLR1, and TRAD gene expression, which are important for activation of nuclear factor-κB (NF-κB) and apoptosis pathways. AAT suppressed expression dose-dependently; 50% inhibition was achieved in the 2.5 to 5.0 mg/mL range. CONCLUSIONS: AAT was well tolerated and safe in subjects with new-onset type 1 diabetes. Weekly doses of AAT greater than 90 mg/kg may be necessary for an optimal therapeutic effect.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , alpha 1-Antitrypsin/therapeutic use , Adolescent , Adult , C-Peptide/blood , Child , Diabetes Mellitus, Type 1/blood , Female , Glycated Hemoglobin/metabolism , Humans , Infusions, Intravenous , Male , Young Adult , alpha 1-Antitrypsin/pharmacokinetics
5.
Nat Commun ; 8(1): 951, 2017 10 16.
Article in English | MEDLINE | ID: mdl-29038423

ABSTRACT

Phosphatidylinositol-3-kinases (PI3K) γ and δ are preferentially enriched in leukocytes, and defects in these signaling pathways have been shown to impair T cell activation. The effects of PI3Kγ and PI3Kδ on alloimmunity remain underexplored. Here, we show that both PI3Kγ -/- and PI3Kδ D910A/D910A mice receiving heart allografts have suppression of alloreactive T effector cells and delayed acute rejection. However, PI3Kδ mutation also dampens regulatory T cells (Treg). After treatment with low dose CTLA4-Ig, PI3Kγ -/- , but not PI3Κδ D910A/D910A , recipients exhibit indefinite prolongation of heart allograft survival. PI3Kδ D910A/D910A Tregs have increased apoptosis and impaired survival. Selective inhibition of PI3Kγ and PI3Kδ (using PI3Kδ and dual PI3Kγδ chemical inhibitors) shows that PI3Kγ inhibition compensates for the negative effect of PI3Kδ inhibition on long-term allograft survival. These data serve as a basis for future PI3K-based immune therapies for transplantation.Phosphatidylinositol-3-kinases (PI3K) γ and δ are key regulators of T cell signaling. Here the author show, using mouse heart allograft transplantation models, that PI3Kγ or PI3Kδ deficiency prolongs graft survival, but selective inhibition of PI3Kγ or PI3Kδ reveals alternative transplant survival outcomes post CTLA4-Ig treatment.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/immunology , Graft Rejection/immunology , Graft Survival/immunology , Heart Transplantation , Lymphocyte Activation/immunology , Phosphatidylinositol 3-Kinases/immunology , Skin Transplantation , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Abatacept/pharmacology , Animals , Class I Phosphatidylinositol 3-Kinases , Class Ib Phosphatidylinositol 3-Kinase/genetics , Gene Knockdown Techniques , Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Models, Animal , Mutation , Phosphatidylinositol 3-Kinases/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Transplantation Tolerance/drug effects
6.
PLoS One ; 11(3): e0150792, 2016.
Article in English | MEDLINE | ID: mdl-26943809

ABSTRACT

The innate immune system critically shapes diabetogenic adaptive immunity during type 1 diabetes (T1D) pathogenesis. While the role of tissue-infiltrating monocyte-derived macrophages in T1D is well established, the role of their tissue-resident counterparts remains undefined. We now demonstrate that islet resident macrophages (IRMs) from non-autoimmune mice have an immunoregulatory phenotype and powerfully induce FoxP3+ Tregs in vitro. The immunoregulatory phenotype and function of IRMs is compromised by TLR4 activation in vitro. Moreover, as T1D approaches in NOD mice, the immunoregulatory phenotype of IRMs is diminished as is their relative abundance compared to immunostimulatory DCs. Our findings suggest that maintenance of IRM abundance and their immunoregulatory phenotype may constitute a novel therapeutic strategy to prevent and/or cure T1D.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/pathology , Macrophages/immunology , Animals , Antigens, CD/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Forkhead Transcription Factors/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Phenotype , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 4/metabolism
7.
Curr Opin Organ Transplant ; 20(1): 8-12, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25563986

ABSTRACT

PURPOSE OF REVIEW: To update knowledge concerning the cause and consequences of the detrimental forms of innate immunity that inevitably occurs in peritransplant period tissue and cellular transplants. In addition, we review the information that a newly discovered, engraftment-promoting, and tolerance-inducing macrophage population is identified and characterized. RECENT FINDINGS: The allograft response mounted by adaptive immune cells is shaped by innate immunity. The early allograft response is uniquely intense as a result of activation of the innate immune response created by ischemia reperfusion injury in organ transplants, delayed revascularization of cell transplants, and hypoxia. Inflammation is created by both cellular 'debris' and cytokines. However, a newly discovered prominent, albeit fragile, tissue-resident, noninvasive, and immunoregulatory macrophage promotes engraftment and tolerance. The role of intracellular 'debris' as well as inflammation in evoking detrimental rejection-provoking peritransplant inflammation is emphasized as well as characterization of a prominent and highly immunoregulatory albeit fragile macrophage population that is tissue-resident and does not circulate is characterized. SUMMARY: Opportunity lies in the ability to rein in detrimental peri-transplant inflammation and in the ability to promote the longevity of a subpopulation of highly potent tissue-resident immunoregulatory macrophages.


Subject(s)
Immune Tolerance/immunology , Immunity, Innate/immunology , Organ Transplantation , Allografts , Animals , Humans
8.
Article in English | MEDLINE | ID: mdl-29942371

ABSTRACT

Leukaemia inhibitory factor (LIF) plays a critical role in "stemness" versus "differentiation", a property that underpins the core value of LIF as a therapeutic for both the treatment of autoimmune disease and for promoting tissue repair. This value can be realized using nano-engineering technology, where a new generation of tools can, with unprecedented ability, manipulate biological functions. One striking example is the treatment of multiple sclerosis (MS). The underpinning biology is the newly identified LIF/IL-6 axis in T lymphocytes, which can tilt the behaviour between immune tolerance versus immune attack. This LIF/IL-6 axis is ideally suited to nanotherapeutic manipulation, given its inherent mechanistic simplicity of two mutually opposing feed-forward loops that determine either tolerogenic (LIF) or inflammatory (IL-6) immunity. Using LIF that is formulated in biodegradable nanoparticles (LIF-NP) and targeted to CD4+ T cells, the axis is harnessed towards immune tolerance. This has implications for the treatment of autoimmune diseases, where the clinical burden is immense. It encompasses more than 100 diseases and, in the USA alone, costs more than $100 billion in direct health care costs annually. Other properties of LIF include the promotion of healthy neuro-glial interactions within the central nervous system (CNS), where, in addition to MS, LIF-NP therapy is relevant to inflammatory neurodegenerative diseases that represent a large and increasing need within aging populations. Thirdly, LIF is a reparative growth factor that can maintain genomic plasticity. LIF-NP supports the use of stem cell-based therapies in regenerative medicine plus augment therapeutic benefits within the patient. These core properties of LIF are greatly amplified in value by the advantage of being formulated as nanoparticles, namely (i) targeted delivery, (ii) exploitation of endogenous regulatory pathways and (iii) creation of surrogate micro-stromal niches. We discuss LIF-NP as a means to harness endogenous pathways for the treatment of MS, both to reset immune self-tolerance and to promote repair of myelin that is required to support health within the nervous system.

9.
Curr Opin Organ Transplant ; 19(6): 545-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25373855

ABSTRACT

PURPOSE OF REVIEW: The field of vascularized composite allograft (VCA) to achieve its full potential will require induction of tolerance. This review will introduce a new method of potential inducing tolerance in hand transplantation. RECENT FINDINGS: Hand transplantation is never a life-extending transplant. This fact resulted in considerable debate both for and against the use of immunosuppression for nonlife-extending transplants. There is considerable debate about the ethics of hand transplantation. There is now consensus that nonlife-extending transplants are acceptable in properly selected patients. However, ideally, hand transplants should not receive life-long immunosuppression. Therefore, attempts to achieve drug-free tolerance through nonlife-endangering therapies are warranted. To this end, we propose implementation of tolerizing therapy long after periinflammation has subsided and drug minimization has proven successful. Evidence that short-term treatment with low doses of IL-2 or a long-lived IL-2 immunoglobulin (Ig) can tilt the balance of immunity from tissue destructive to tolerance come from preclinical demonstrations in mouse and nonhuman primate models of autoimmunity and/or transplantation and even more recent clinical trials. SUMMARY: We believe that with the proper use of low-dose IL-2 given at an opportune time in the inflammatory process of transplant that reduce immunosuppression and even tolerance can be induced in hand transplantation. We propose that tolerance can be inducted after a long period of conventional treatment to avoid 'tolerance-hindering' adverse inflammation that occurs in the posttransplant period. With abatement of posttransplant inflammation and with time, we will institute low-dose IL-2-based therapy to support the proliferation, viability and functional phenotype of regulatory T cells.


Subject(s)
Hand Transplantation , Immune Tolerance , Animals , Graft Rejection/immunology , Humans , Immunologic Memory , Inflammation/immunology , Interleukin-2/immunology
10.
Adv Exp Med Biol ; 809: 103-16, 2014.
Article in English | MEDLINE | ID: mdl-25302368

ABSTRACT

Effective means to identify anti-donor immune activity before the transplant organ is damaged and rejected has been an important goal in transplantation research. Development of sensitive and non-invasive diagnostic methods that probe the immune status of the recipient as well as the resilience of the donor organ should enable personalized application of immunosuppressive drugs. With a non-invasive biomarker for rejection, it should be possible to selectively treat the patients that are rejecting the graft and wean the tolerant patients from immunosuppression. Although A20 is also expressed by activated CD4+ T cells and CD8+ T cells, its expression by mouse tubular cells has been shown to play an important role in protecting allografts from ischemia/reperfusion (I/R) injury and rejection. Using quantitative (real-time) reverse transcriptase polymerase chain reaction (qt-RT-PCR), we showed that expression levels of A20, heme oxygenase (HO)-1, other anti-apoptotic molecules, granzyme-B (GZMB), perforin (PRF1), CD3 and other immune molecules in renal transplant biopsies, urinary cells and peripheral blood cells are predictive of transplantation outcomes. Measuring A20 at mRNA and protein levels has the potentiality to be diagnostic and prognostic of transplantation outcomes and thereby help in timely therapeutic interventions to prolong graft life.


Subject(s)
Biomarkers/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Transplantation , Nuclear Proteins/metabolism , Apoptosis , Humans , Monitoring, Physiologic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Transplantation, Homologous , Tumor Necrosis Factor alpha-Induced Protein 3
11.
Curr Opin Organ Transplant ; 19(6): 531-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25333831

ABSTRACT

PURPOSE OF REVIEW: Acute rejection is the most common complication after vascularized composite allotransplantation (VCA). This review provides a state-of-the-art analysis of prevention, diagnosis and treatment of acute rejection episodes and highlights recent findings with the potential to improve patient care and enhance understanding of the underlying biologic processes. RECENT FINDINGS: Recent reports suggest that maintenance immunosuppression dose reduction and steroid withdrawal are realistic goals in VCA, despite the known high immunogenicity of the skin component. It appears that utilization of sentinel flaps, in-depth histological analyses and application of novel biomarkers have facilitated early diagnosis and characterization of acute rejection episodes, leading to timely institution of appropriate therapy. The successful management of the first highly sensitized face transplant recipient suggests the possibility of carefully considering these high-risk VCA candidates for transplantation. SUMMARY: Acute rejection is higher in VCA than in any other organ in the field of transplantation, although most episodes are controlled by high-dose steroids and optimization of maintenance immunosuppression. Because of limitations in patient number and the duration of follow-up, the long-term safety and effectiveness of VCA remain unclear. Moreover, the tests currently used to diagnose acute rejection are of limited value. Better diagnostic tools and a better understanding of the immunologic events during acute rejection are therefore needed to improve diagnosis, treatment and outcomes of this life-changing restorative surgery.


Subject(s)
Graft Rejection/immunology , Acute Disease , Animals , Graft Rejection/prevention & control , Graft Rejection/therapy , Humans , Immune Tolerance , Skin/immunology , Skin Transplantation , Vascularized Composite Allotransplantation
12.
J Clin Invest ; 124(8): 3443-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24983317

ABSTRACT

Macrophages characterized as M2 and M2-like regulate immune responses associated with immune suppression and healing; however, the relationship of this macrophage subset to CD169+ tissue-resident macrophages and their contribution to shaping alloimmune responses is unknown. Here we identified a population of M2-like tissue-resident macrophages that express high levels of the phosphatidylserine receptor TIM-4 and CD169 (TIM-4hiCD169+). Labeling and tracking of TIM-4hiCD169+ macrophages in mice revealed that this population is a major subset of tissue-resident macrophages, homes to draining LNs following oxidative stress, exhibits an immunoregulatory and hypostimulatory phenotype that is maintained after migration to secondary lymphoid organs, favors preferential induction of antigen-stimulated Tregs, and is highly susceptible to apoptosis. Moreover, CD169+ tissue-resident macrophages were resistant to oxidative stress-induced apoptosis in mice lacking TIM-4. Compared with heart allografts from WT mice, Tim4-/- heart allografts survived much longer and were more easily tolerized by non-immunosuppressed recipients. Furthermore, Tim4-/- allograft survival was associated with the infiltration of Tregs into the graft. Together, our data provide evidence that M2-like TIM-4hiCD169+ tissue-resident macrophages are immunoregulatory and promote engraftment of cardiac allografts, but their influence is diminished by TIM-4-dependent programmed cell death.


Subject(s)
Macrophages/immunology , Macrophages/physiology , Membrane Proteins/metabolism , Allografts , Animals , Apoptosis , Cell Differentiation , Cell Movement , Cell Proliferation , Graft Survival , Heart Transplantation , Lymphocyte Culture Test, Mixed , Macrophages/cytology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidative Stress , Sialic Acid Binding Ig-like Lectin 1/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Transplantation Tolerance
13.
Clin Immunol ; 153(1): 8-16, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24691417

ABSTRACT

Harness of sensitized transplantation remains a clinical challenge particularly in parallel with prolonged cold ischemia time (PCI)-mediated injury. Our present study was to test the role of myeloid-derived suppressor cells (MDSCs) in mouse pre-sensitized transplantation. Our findings revealed that CD11b+Gr1(low) MDSC was shown to have strong suppressive activity. MDSCs subsets from the tolerated mice exhibited higher suppressive capacities compared with counterparts from naive (untreated) mice. Depletion of Tregs could not affect splenic CD11b+Gr1(-low) MDSC frequency, but increase peripheral and intragraft CD11b+Gr1(-low) frequency. Intriguingly, boost of Tregs remarkably caused an increase of CD11b+Gr1(-low) frequency in the graft, peripheral blood, and spleen. Furthermore, peripheral CD11b+Gr1(-low) cells were massively accumulated at the early stage when allogeneic immune response was enhanced. Taken together, MDSCs could prevent grafts from PCI-mediated injury independent on Tregs in the pre-sensitized transplant recipients. Utilization of MDSC subset particularly CD11b+Gr1(-low) might provide a novel insight into improving graft outcome under such clinical scenarios.


Subject(s)
Heart Transplantation , Myeloid Cells/immunology , Animals , CD11b Antigen/metabolism , Cell Count , Cold Ischemia/adverse effects , Disease Models, Animal , Graft Survival/immunology , Immunophenotyping , Immunosuppression Therapy , Male , Mice , Myeloid Cells/metabolism , Receptors, Chemokine/metabolism , Skin Transplantation , Spleen/cytology , Spleen/immunology
14.
Article in English | MEDLINE | ID: mdl-24370928

ABSTRACT

We are entering an exciting time in the study of immunologic tolerance. Several cellular and molecular strategies have been developed that show promise in nonhuman transplant models and these approaches are just now appearing in clinical trials. Tolerance strategies that prevent immune rejection and obviate the need for immunosuppressive medications (with inherent risk of cancer, infection, and organ toxicity) would improve both graft and patient survival. Each tolerance protocol brings its own set of associated risks. As the results of these trials become available, we must continue to evaluate their successes and failures. The balance of these outcomes will help us answer the question: "Tolerance-Is it worth it?"


Subject(s)
Graft Rejection/immunology , Immunosuppression Therapy/methods , Immunosuppressive Agents/adverse effects , Transplantation Tolerance/immunology , Costimulatory and Inhibitory T-Cell Receptors/antagonists & inhibitors , Costs and Cost Analysis , Graft Rejection/genetics , Humans , Immunosuppression Therapy/adverse effects , Stem Cell Transplantation/adverse effects , T-Lymphocytes, Regulatory/immunology , Transplantation Chimera/immunology , Transplantation Conditioning/methods , Transplantation Tolerance/drug effects
15.
Eur J Immunol ; 44(3): 794-806, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24271843

ABSTRACT

Leptin is an adipose-secreted hormone that plays an important role in both metabolism and immunity. Leptin has been shown to induce Th1-cell polarization and inhibit Th2-cell responses. Additionally, leptin induces Th17-cell responses, inhibits regulatory T (Treg) cells and modulates autoimmune diseases. Here, we investigated whether leptin mediates its activity on T cells by influencing dendritic cells (DCs) to promote Th17 and Treg-cell immune responses in mice. We observed that leptin deficiency (i) reduced the expression of DC maturation markers, (ii) decreased DC production of IL-12, TNF-α, and IL-6, (iii) increased DC production of TGF-ß, and (iv) limited the capacity of DCs to induce syngeneic CD4(+) T-cell proliferation. As a consequence of this unique phenotype, DCs generated under leptin-free conditions induced Treg or TH 17 cells more efficiently than DCs generated in the presence of leptin. These data indicate important roles for leptin in DC homeostasis and the initiation and maintenance of inflammatory and regulatory immune responses by DCs.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Leptin/deficiency , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Animals , Dendritic Cells/immunology , Immunophenotyping , Leptin/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Phenotype , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/cytology , Th17 Cells/immunology
16.
Nat Rev Nephrol ; 9(11): 631-2, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24080796

ABSTRACT

Renal biopsy is the gold standard for detection of rejection in kidney transplant recipients, but is not considered until evidence of renal dysfunction is apparent. Now, Suthanthiran and colleagues suggest that mRNA levels in urinary cells from these patients might be diagnostic and prognostic of acute cellular rejection.


Subject(s)
Graft Rejection/genetics , Graft Rejection/pathology , Kidney Transplantation/adverse effects , RNA, Messenger/metabolism , Acute Disease , Biopsy , Humans , Prognosis
17.
PLoS One ; 8(8): e69257, 2013.
Article in English | MEDLINE | ID: mdl-23990881

ABSTRACT

We describe a novel photoconversion technique to track individual cells in vivo using a commercial lipophilic membrane dye, DiR. We show that DiR exhibits a permanent fluorescence emission shift (photoconversion) after light exposure and does not reacquire the original color over time. Ratiometric imaging can be used to distinguish photoconverted from non-converted cells with high sensitivity. Combining the use of this photoconvertible dye with intravital microscopy, we tracked the division of individual hematopoietic stem/progenitor cells within the calvarium bone marrow of live mice. We also studied the peripheral differentiation of individual T cells by tracking the gain or loss of FoxP3-GFP expression, a marker of the immune suppressive function of CD4(+) T cells. With the near-infrared photoconvertible membrane dye, the entire visible spectral range is available for simultaneous use with other fluorescent proteins to monitor gene expression or to trace cell lineage commitment in vivo with high spatial and temporal resolution.


Subject(s)
Cell Membrane/metabolism , Coloring Agents/chemistry , Photochemistry , Single-Cell Analysis/methods , Animals , Bone Marrow/metabolism , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Lineage , Forkhead Transcription Factors/metabolism , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Spectrometry, Fluorescence , Staining and Labeling/methods , Stem Cells/cytology , T-Lymphocytes/cytology , Time Factors
18.
J Clin Invest ; 123(6): 2360-2, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23676457

ABSTRACT

During transplant rejection, migrating T cells infiltrate the grafted organ, but the signals that direct this migration are incompletely understood. In this issue of the JCI, Walch et al. debunk two classical paradigms concerning transplant rejection, with important consequences for the design of antirejection therapeutics.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/physiology , Heart Transplantation/immunology , Kidney Transplantation/immunology , Transendothelial and Transepithelial Migration/immunology , Animals
19.
Mol Pharm ; 10(2): 717-27, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23311475

ABSTRACT

Fc fusion proteins are a new emerging class of molecules for immune-targeted delivery of therapeutic proteins. Biophysical and bioanalytical characterization is critical for clinical development and delivery of therapeutic proteins. Here we report molecular and functional characterization of a recombinant human fusion protein Mutant IL-15/Fc. MutIL-15/Fc has a molecular weight of ∼95 kDa as determined by multiangle laser light scattering with online size exclusion chromatography and migrated at a faster rate (lower retention time) in gel filtration column. The kinetics of binding of MutIL-15/Fc to Fcγ receptor is best fitted in a bivalent modal with K(D1) 5 µM and K(D2) 9 µM determined by surface plasmon resonance (BIAcore). N-Glycoprofiling analysis revealed extensive glycosylation of MutIL-15/Fc. The Fc and IL-15 components in the MutIL-15/Fc are detected using the dual mode ELISA. The HT-2 cell proliferation inhibition assay is qualified as a quantitative in vitro marker functional assay. Molecular state changes associated with forced stress analyzed by SEC-MALS resulted in changes in bioactivity and Fc:Fcγ receptor interaction affinity. These data provide a systematic approach to molecular and functional characterization of the MutIL-15/Fc to establish product consistency and stability monitoring during storage and under drug delivery conditions.


Subject(s)
Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Interleukin-15/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Cell Proliferation , Chromatography, Gel , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fc Fragments/genetics , Kinetics , Protein Binding , Receptors, IgG/metabolism
20.
Sci Signal ; 5(254): ra90, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23233528

ABSTRACT

T cell immunoglobulin and mucin domain (TIM) proteins are cell-surface signaling receptors in T cells and scavenger receptors in antigen-presenting cells and kidney tubular epithelia. Here, we demonstrated a function for TIM proteins in mediating the degradation of NUR77, a nuclear receptor implicated in apoptosis and cell survival. TIM proteins interacted with and mediated the lysosomal degradation of NUR77 in a phosphoinositide 3-kinase-dependent pathway. We also showed dynamic cycling of TIM-1 to and from the cell surface through clathrin-dependent constitutive endocytosis. Blocking this process or mutating the phosphatidylserine-binding pocket in TIM-1 abrogated TIM-1-mediated degradation of NUR77. In an in vitro model of kidney injury, silencing TIM-1 increased NUR77 abundance and decreased epithelial cell survival. These results show that TIM proteins may affect immune cell function and the response of the kidney to injury.


Subject(s)
Lysosomes/metabolism , Membrane Glycoproteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Proteolysis , Receptors, Virus/metabolism , T-Lymphocytes/metabolism , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Endocytosis/physiology , Epithelial Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Hepatitis A Virus Cellular Receptor 1 , Humans , Kidney/cytology , Luciferases , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/physiology , Real-Time Polymerase Chain Reaction , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...