Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Semin Immunopathol ; 45(1): 145-157, 2023 01.
Article in English | MEDLINE | ID: mdl-36414691

ABSTRACT

Multiplexed imaging, which enables spatial localization of proteins and RNA to cells within tissues, complements existing multi-omic technologies and has deepened our understanding of health and disease. CODEX, a multiplexed single-cell imaging technology, utilizes a microfluidics system that incorporates DNA barcoded antibodies to visualize 50 + cellular markers at the single-cell level. Here, we discuss the latest applications of CODEX to studies of cancer, autoimmunity, and infection as well as current bioinformatics approaches for analysis of multiplexed imaging data from preprocessing to cell segmentation and marker quantification to spatial analysis techniques. We conclude with a commentary on the challenges and future developments for multiplexed spatial profiling.


Subject(s)
Computational Biology , Multiomics , Single-Cell Analysis , Humans , Microfluidics , Neoplasms , Autoimmune Diseases , Infections
3.
Proc Natl Acad Sci U S A ; 116(52): 26727-26733, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31822623

ABSTRACT

Foxp3+CD4+ regulatory T cells (Tregs) play important roles in controlling both homeostatic processes and immune responses at the tissue and organismal levels. For example, Tregs promote muscle regeneration in acute or chronic injury models by direct effects on local muscle progenitor cells, as well as on infiltrating inflammatory cells. Muscle Tregs have a transcriptome, a T cell receptor (TCR) repertoire, and effector capabilities distinct from those of classical, lymphoid-organ Tregs, but it has proven difficult to study the provenance and functions of these unique features due to the rarity of muscle Tregs and their fragility on isolation. Here, we attempted to sidestep these hindrances by generating, characterizing, and employing a line of mice carrying rearranged transgenes encoding the TCRα and TCRß chains from a Treg clone rapidly and specifically expanded within acutely injured hindlimb muscle of young mice. Tregs displaying the transgene-encoded TCR preferentially accumulated in injured hindlimb muscle in a TCR-dependent manner both in the straight transgenic model and in adoptive-transfer systems; non-Treg CD4+ T cells expressing the same TCR did not specifically localize in injured muscle. The definitive muscle-Treg transcriptome was not established until the transgenic Tregs inhabited muscle. When crossed onto the mdx model of Duchenne muscular dystrophy, the muscle-Treg TCR transgenes drove enhanced accumulation of Tregs in hindlimb muscles and improved muscle regeneration. These findings invoke the possibility of harnessing muscle Tregs or their TCRs for treatment of skeletal muscle pathologies.

4.
Cell ; 174(2): 285-299.e12, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29887374

ABSTRACT

Visceral adipose tissue (VAT) hosts a population of regulatory T (Treg) cells, with a unique phenotype, that controls local and systemic inflammation and metabolism. Generation of a T cell receptor transgenic mouse line, wherein VAT Tregs are highly enriched, facilitated study of their provenance, dependencies, and activities. We definitively established a role for T cell receptor specificity, uncovered an unexpected function for the primordial Treg transcription-factor, Foxp3, evidenced a cell-intrinsic role for interleukin-33 receptor, and ordered these dependencies within a coherent scenario. Genesis of the VAT-Treg phenotype entailed a priming step in the spleen, permitting them to exit the lymphoid organs and surveil nonlymphoid tissues, and a final diversification process within VAT, in response to microenvironmental cues. Understanding the principles of tissue-Treg biology is a prerequisite for precision-targeting strategies.


Subject(s)
Intra-Abdominal Fat/metabolism , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory/metabolism , Animals , Chromatin Assembly and Disassembly , Forkhead Transcription Factors/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Intra-Abdominal Fat/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , PPAR gamma/genetics , PPAR gamma/metabolism , Phenotype , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin/metabolism , Single-Cell Analysis , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transcriptome
5.
Semin Arthritis Rheum ; 47(6): 907-910, 2018 06.
Article in English | MEDLINE | ID: mdl-29191375

ABSTRACT

OBJECTIVE: Immune checkpoint inhibitors are effective cancer therapies that have been associated with immune-related adverse events (irAEs). Recent reports of irAEs describe symptoms resembling classic rheumatologic syndromes, most notably associated with cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitor blockade. Though cases have been described, there are fewer reports of rheumatologic disease associated with programmed cell death protein-1 (PD-1) inhibitors. Here, we describe a series of four patients presenting to the Brigham and Women's Hospital (BWH) Arthritis Center with de novo polymyalgia rheumatica (PMR)-type conditions and/or peripheral synovitis after treatment with PD-1/PD-Ligand 1 (PD-L1) pathway inhibitors. METHODS: Patients with metastatic renal cell carcinoma (RCC) who were treated with PD-1/PD-L1 pathway inhibitors and subsequently developed complaints of new joint pain were referred to the BWH Arthritis Center as part of routine care and identified retrospectively. The electronic medical record was reviewed for cancer history and treatment, rheumatologic symptoms, physical exam, laboratory testing, and clinical course. RESULTS: All four patients developed irAEs consistent with a PMR-type syndrome and/or peripheral synovitis. Symptoms persisted despite discontinuation of the PD-1/PD-L1 pathway inhibitors; however, three of the patients responded well to oral glucocorticoids alone while one patient required the addition of oral methotrexate. All patients had an eventual decline in inflammatory markers. CONCLUSION: These cases highlight the need for both oncologists and rheumatologists to recognize the development of rheumatologic disease during treatment with immune checkpoint blockade. Further investigation is needed to optimize the management of irAEs, particularly considering the increasing use of checkpoint inhibitors to treat malignancies.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Polymyalgia Rheumatica/chemically induced , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Synovitis/chemically induced , Aged , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Female , Humans , Kidney Neoplasms/drug therapy , Middle Aged , Nivolumab , Retrospective Studies
6.
Immunity ; 44(2): 355-67, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26872699

ABSTRACT

Normal repair of skeletal muscle requires local expansion of a special population of Foxp3(+)CD4(+) regulatory T (Treg) cells. Such cells failed to accumulate in acutely injured muscle of old mice, known to undergo ineffectual repair. This defect reflected reduced recruitment of Treg cells to injured muscle, as well as less proliferation and retention therein. Interleukin-33 (IL-33) regulated muscle Treg cell homeostasis in young mice, and its administration to old mice ameliorated their deficits in Treg cell accumulation and muscle regeneration. The major IL-33-expressing cells in skeletal muscle displayed a constellation of markers diagnostic of fibro/adipogenic progenitor cells and were often associated with neural structures, including nerve fibers, nerve bundles, and muscle spindles, which are stretch-sensitive mechanoreceptors important for proprioception. IL-33(+) cells were more frequent after muscle injury and were reduced in old mice. IL-33 is well situated to relay signals between the nervous and immune systems within the muscle context.


Subject(s)
Aging/immunology , Interleukin-33/metabolism , Muscle, Skeletal/physiology , Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Adipogenesis/immunology , Animals , Biomarkers/metabolism , Cell Proliferation , Cells, Cultured , Homeostasis , Humans , Mechanotransduction, Cellular , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/innervation , Nerve Fibers/physiology , Neuroimmunomodulation , Regeneration/immunology , Wound Healing
7.
Cell ; 155(6): 1282-95, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24315098

ABSTRACT

Long recognized to be potent suppressors of immune responses, Foxp3(+)CD4(+) regulatory T (Treg) cells are being rediscovered as regulators of nonimmunological processes. We describe a phenotypically and functionally distinct population of Treg cells that rapidly accumulated in the acutely injured skeletal muscle of mice, just as invading myeloid-lineage cells switched from a proinflammatory to a proregenerative state. A Treg population of similar phenotype accumulated in muscles of genetically dystrophic mice. Punctual depletion of Treg cells during the repair process prolonged the proinflammatory infiltrate and impaired muscle repair, while treatments that increased or decreased Treg activities diminished or enhanced (respectively) muscle damage in a dystrophy model. Muscle Treg cells expressed the growth factor Amphiregulin, which acted directly on muscle satellite cells in vitro and improved muscle repair in vivo. Thus, Treg cells and their products may provide new therapeutic opportunities for wound repair and muscular dystrophies.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Regeneration , T-Lymphocytes, Regulatory/physiology , Amphiregulin , Animals , EGF Family of Proteins , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphoid Tissue/cytology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/immunology , Muscle, Skeletal/injuries , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , Muscular Dystrophies/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transcriptome
8.
Immunity ; 38(2): 296-308, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23333075

ABSTRACT

Macrophages frequently infiltrate tumors and can enhance cancer growth, yet the origins of the macrophage response are not well understood. Here we address molecular mechanisms of macrophage production in a conditional mouse model of lung adenocarcinoma. We report that overproduction of the peptide hormone Angiotensin II (AngII) in tumor-bearing mice amplifies self-renewing hematopoietic stem cells (HSCs) and macrophage progenitors. The process occurred in the spleen but not the bone marrow, and was independent of hemodynamic changes. The effects of AngII required direct hormone ligation on HSCs, depended on S1P(1) signaling, and allowed the extramedullary tissue to supply new tumor-associated macrophages throughout cancer progression. Conversely, blocking AngII production prevented cancer-induced HSC and macrophage progenitor amplification and thus restrained the macrophage response at its source. These findings indicate that AngII acts upstream of a potent macrophage amplification program and that tumors can remotely exploit the hormone's pathway to stimulate cancer-promoting immunity.


Subject(s)
Adenocarcinoma/metabolism , Angiotensin II/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Spleen/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Angiotensin II/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Communication , Cell Movement , Cell Proliferation , Gene Expression , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lysophospholipids/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Spleen/pathology , Tumor Burden
9.
Proc Natl Acad Sci U S A ; 109(31): 12668-73, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22733744

ABSTRACT

To protect the organism against autoimmunity, self-reactive effector/memory T cells (T(E/M)) are controlled by cell-intrinsic and -extrinsic regulatory mechanisms. However, how some T(E/M) cells escape regulation and cause autoimmune disease is currently not understood. Here we show that blocking IL-7 receptor-α (IL-7Rα) with monoclonal antibodies in nonobese diabetic (NOD) mice prevented autoimmune diabetes and, importantly, reversed disease in new-onset diabetic mice. Surprisingly, IL-7-deprived diabetogenic T(E/M) cells remained present in the treated animals but showed increased expression of the inhibitory receptor Programmed Death 1 (PD-1) and reduced IFN-γ production. Conversely, IL-7 suppressed PD-1 expression on activated T cells in vitro. Adoptive transfer experiments revealed that T(E/M) cells from anti-IL-7Rα-treated mice had lost their pathogenic potential, indicating that absence of IL-7 signals induces cell-intrinsic tolerance. In addition to this mechanism, IL-7Rα blockade altered the balance of regulatory T cells and T(E/M) cells, hence promoting cell-extrinsic regulation and further increasing the threshold for diabetogenic T-cell activation. Our data demonstrate that IL-7 contributes to the pathogenesis of autoimmune diabetes by enabling T(E/M) cells to remain in a functionally competent state and suggest IL-7Rα blockade as a therapy for established T-cell-dependent autoimmune diseases.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Immunologic Memory , Interleukin-7/immunology , Receptors, Interleukin-7/antagonists & inhibitors , Signal Transduction/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antigens, Differentiation/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Female , Immune Tolerance , Interferon-gamma/immunology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Programmed Cell Death 1 Receptor , Receptors, Interleukin-7/immunology , T-Lymphocytes/pathology
10.
J Immunol ; 185(11): 6426-30, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21037099

ABSTRACT

IL-2 controls the survival of regulatory T cells (Tregs), but it is unclear whether IL-2 also directly affects Treg suppressive capacity in vivo. We have found that eliminating Bim-dependent apoptosis in IL-2- and CD25-deficient mice restored Treg numbers but failed to cure their lethal autoimmune disease, demonstrating that IL-2-dependent survival and suppressive activity can be uncoupled in Tregs. Treatment with IL-2-anti-IL-2-Ab complexes enhanced the numbers and suppressive capacity of IL-2-deprived Tregs with striking increases in CD25, CTLA-4, and CD39/CD73 expression. Although cytokine treatment induced these suppressive mechanisms in both IL-2(-/-) and IL-2(-/-)Bim(-/-) mice, it only reversed autoimmune disease in the latter. Our results suggest that successful IL-2 therapy of established autoimmune diseases will require a threshold quantity of Tregs present at the start of treatment and show that the suppressive capacity of Tregs critically depends on IL-2 even when Treg survival is independent of this cytokine.


Subject(s)
Interleukin-2/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Anemia, Hemolytic, Autoimmune/genetics , Anemia, Hemolytic, Autoimmune/immunology , Anemia, Hemolytic, Autoimmune/therapy , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Proliferation , Cells, Cultured , Coculture Techniques , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Deletion , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/deficiency , Interleukin-2 Receptor alpha Subunit/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Regulatory/pathology
11.
J Immunol ; 183(1): 332-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542444

ABSTRACT

An early reaction of CD4(+) T lymphocytes to Ag is the production of cytokines, notably IL-2. To detect cytokine-dependent responses, naive Ag-specific T cells were stimulated in vivo and the presence of phosphorylated STAT5 molecules was used to identify the cell populations responding to IL-2. Within hours of T cell priming, IL-2-dependent STAT5 phosphorylation occurred primarily in Foxp3(+) regulatory T cells. In contrast, the Ag-specific T cells received STAT5 signals only after repeated Ag exposure or memory differentiation. Regulatory T cells receiving IL-2 signals proliferated and developed enhanced suppressive activity. These results indicate that one of the earliest events in a T cell response is the activation of endogenous regulatory cells, potentially to prevent autoimmunity.


Subject(s)
Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Proliferation , Interleukin-2/biosynthesis , Interleukin-2/physiology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Phosphorylation , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , T-Lymphocytes, Regulatory/virology , Time Factors , Vaccinia virus/immunology
12.
Blood ; 113(2): 389-95, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-18815283

ABSTRACT

Imbalance of T-helper cell (Th) differentiation and subsequent cytokine dysregulation is implicated in inflammatory and autoimmune diseases. In particular, 2 cytokines produced by different Th cell populations, interferon-gamma (IFN-gamma) and interleukin-17 (IL-17), have been shown to play a critical role in autoimmunity. We have examined the roles of these cytokines in a mouse model of systemic autoimmunity resulting from the deletion of IL-2 in which autoimmune hemolytic anemia (AIHA) is a prominent feature. We demonstrate that, in IL-2-knockout (KO) BALB/c mice, elimination of the Th1 cytokine, IFN-gamma, delays the development of AIHA. Further, CD4(+) T cells from IL-2/IFN-gamma-KO mice produce elevated levels of IL-17 compared with wild-type (WT) and IL-2-KO, and these mice eventually develop intestinal inflammation. In contrast, elimination of the Th17 cytokine, IL-17, from IL-2-KO mice fails to suppress early acute AIHA development. These results suggest that in a systemic autoimmune disease with multiple manifestations, Th1 cells drive the early autoantibody response and IL-17-producing cells may be responsible for the more chronic tissue inflammation.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , Autoimmunity , Cytokines/immunology , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Acute Disease , Anemia, Hemolytic, Autoimmune/genetics , Anemia, Hemolytic, Autoimmune/metabolism , Anemia, Hemolytic, Autoimmune/pathology , Animals , Autoimmunity/genetics , Chronic Disease , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Th1 Cells/metabolism , Th1 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...