Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Sci Immunol ; 8(85): eadd1591, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37506196

ABSTRACT

Immune checkpoint inhibitor (ICI) therapies used to treat cancer, such as anti-PD-1 antibodies, can induce autoimmune conditions in some individuals. The T cell mechanisms mediating such iatrogenic autoimmunity and their overlap with spontaneous autoimmune diseases remain unclear. Here, we compared T cells from the joints of 20 patients with an inflammatory arthritis induced by ICI therapy (ICI-arthritis) with two archetypal autoimmune arthritides, rheumatoid arthritis (RA) and psoriatic arthritis (PsA). Single-cell transcriptomic and antigen receptor repertoire analyses highlighted clonal expansion of an activated effector CD8 T cell population in the joints and blood of patients with ICI-arthritis. These cells were identified as CD38hiCD127- CD8 T cells and were uniquely enriched in ICI-arthritis joints compared with RA and PsA and also displayed an elevated interferon signature. In vitro, type I interferon induced CD8 T cells to acquire the ICI-associated CD38hi phenotype and enhanced cytotoxic function. In a cohort of patients with advanced melanoma, ICI therapy markedly expanded circulating CD38hiCD127- T cells, which were frequently bound by the therapeutic anti-PD-1 drug. In patients with ICI-arthritis, drug-bound CD8 T cells in circulation showed marked clonal overlap with drug-bound CD8 T cells from synovial fluid. These results suggest that ICI therapy directly targets CD8 T cells in patients who develop ICI-arthritis and induces an autoimmune pathology that is distinct from prototypical spontaneous autoimmune arthritides.


Subject(s)
Arthritis, Psoriatic , Arthritis, Rheumatoid , CD8-Positive T-Lymphocytes , Humans , Arthritis, Psoriatic/metabolism , Synovial Fluid/metabolism , T-Lymphocytes, Cytotoxic/metabolism
2.
Curr Opin Rheumatol ; 35(4): 249-254, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37040654

ABSTRACT

PURPOSE OF REVIEW: To summarize recently discovered novel cell states in rheumatoid arthritis (RA) synovium that could have important implications for disease treatment. RECENT FINDINGS: The use of multiomic technologies, including single-cell and spatial transcriptomics and mass cytometry, has led to the discovery of several novel cell states, which could have important implications for the treatment of RA. These cells can be found in patient blood, synovial fluid, or synovial tissue and span several immune cell subsets as well as stromal cell types. These diverse cell states may represent the targets of current or future therapeutics, while their fluctuations may inform the ideal timing for therapy. Future efforts are needed to implicate how each cell state functions in the pathophysiologic network within affected joints and how medications perturb each cell state and ultimately the tissue. SUMMARY: Multiomic molecular technologies have afforded the discovery of numerous novel cellular states in RA synovium; the next challenge will be to link these states to pathophysiology and treatment response.


Subject(s)
Arthritis, Rheumatoid , Humans , Synovial Membrane/metabolism , Synovial Fluid , Stromal Cells
3.
Cell Stem Cell ; 28(11): 1922-1935.e5, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34529935

ABSTRACT

Little is known about how interactions of diet, intestinal stem cells (ISCs), and immune cells affect early-stage intestinal tumorigenesis. We show that a high-fat diet (HFD) reduces the expression of the major histocompatibility complex class II (MHC class II) genes in intestinal epithelial cells, including ISCs. This decline in epithelial MHC class II expression in a HFD correlates with reduced intestinal microbiome diversity. Microbial community transfer experiments suggest that epithelial MHC class II expression is regulated by intestinal flora. Mechanistically, pattern recognition receptor (PRR) and interferon-gamma (IFNγ) signaling regulates epithelial MHC class II expression. MHC class II-negative (MHC-II-) ISCs exhibit greater tumor-initiating capacity than their MHC class II-positive (MHC-II+) counterparts upon loss of the tumor suppressor Apc coupled with a HFD, suggesting a role for epithelial MHC class II-mediated immune surveillance in suppressing tumorigenesis. ISC-specific genetic ablation of MHC class II increases tumor burden cell autonomously. Thus, HFD perturbs a microbiome-stem cell-immune cell interaction that contributes to tumor initiation in the intestine.


Subject(s)
Histocompatibility Antigens Class II , Intestines , Carcinogenesis , Diet, High-Fat , Epithelial Cells , Humans
4.
Bio Protoc ; 11(11): e4012, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34250200

ABSTRACT

Flow cytometry is a popular laser-based technology that allows the phenotypic and functional characterization of individual cells in a high-throughput manner. Here, we describe a detailed procedure for preparing a single-cell suspension from mammary tumors of the mouse mammary tumor virus-polyoma middle T (MMTV-PyMT) and analyzing these cells by multi-color flow cytometry. This protocol can be used to study the following tumor-infiltrating immune cell populations, defined by the expression of cell surface molecules: total leukocytes, tumor-associated macrophages (TAMs), conventional dendritic cells (DCs), CD103-expressing DCs, tumor-associated neutrophils, inflammatory monocytes, natural killer (NK) cells, CD4+ T cells, CD8+ T cells, γδT cells, and regulatory T cells.

5.
J Exp Med ; 217(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32667673

ABSTRACT

C-C chemokine receptor type 2 (CCR2) is expressed on monocytes and facilitates their recruitment to tumors. Though breast cancer cells also express CCR2, its functions in these cells are unclear. We found that Ccr2 deletion in cancer cells led to reduced tumor growth and approximately twofold longer survival in an orthotopic, isograft breast cancer mouse model. Deletion of Ccr2 in cancer cells resulted in multiple alterations associated with better immune control: increased infiltration and activation of cytotoxic T lymphocytes (CTLs) and CD103+ cross-presenting dendritic cells (DCs), as well as up-regulation of MHC class I and down-regulation of checkpoint regulator PD-L1 on the cancer cells. Pharmacological or genetic targeting of CCR2 increased cancer cell sensitivity to CTLs and enabled the cancer cells to induce DC maturation toward the CD103+ subtype. Consistently, Ccr2-/- cancer cells did not induce immune suppression in Batf3-/- mice lacking CD103+ DCs. Our results establish that CCR2 signaling in cancer cells can orchestrate suppression of the immune response.


Subject(s)
Adaptive Immunity/immunology , Immune Tolerance , Mammary Neoplasms, Experimental/immunology , Receptors, CCR2/physiology , Adaptive Immunity/physiology , Animals , Apoptosis , B7-H1 Antigen/metabolism , Dendritic Cells/immunology , Dendritic Cells/physiology , Female , Histocompatibility Antigens Class I/metabolism , Immune Tolerance/immunology , Immune Tolerance/physiology , Interferons/metabolism , Mice , Mice, Inbred C57BL , Receptors, CCR2/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/physiology
7.
Sci Transl Med ; 8(361): 361ra138, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27798263

ABSTRACT

Neutrophils, the most abundant type of leukocytes in blood, can form neutrophil extracellular traps (NETs). These are pathogen-trapping structures generated by expulsion of the neutrophil's DNA with associated proteolytic enzymes. NETs produced by infection can promote cancer metastasis. We show that metastatic breast cancer cells can induce neutrophils to form metastasis-supporting NETs in the absence of infection. Using intravital imaging, we observed NET-like structures around metastatic 4T1 cancer cells that had reached the lungs of mice. We also found NETs in clinical samples of triple-negative human breast cancer. The formation of NETs stimulated the invasion and migration of breast cancer cells in vitro. Inhibiting NET formation or digesting NETs with deoxyribonuclease I (DNase I) blocked these processes. Treatment with NET-digesting, DNase I-coated nanoparticles markedly reduced lung metastases in mice. Our data suggest that induction of NETs by cancer cells is a previously unidentified metastasis-promoting tumor-host interaction and a potential therapeutic target.


Subject(s)
Extracellular Traps , Neoplasm Metastasis , Neutrophils/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Deoxyribonuclease I/chemistry , Humans , Lung/pathology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neutrophils/cytology
8.
Dis Model Mech ; 6(3): 580-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23616077

ABSTRACT

The prognosis of metastatic cancer in patients is poor. Interfering with metastatic spread is therefore important for achieving better survival from cancer. Metastatic disease is established through a series of steps, including breaching of the basement membrane, intravasation and survival in lymphatic or blood vessels, extravasation, and growth at distant sites. Yet, although we know the steps involved in metastasis, the cellular and molecular mechanisms of dissemination and colonization of distant organs are incompletely understood. Here, we review the important insights into the metastatic process that have been gained specifically through the use of imaging technologies in murine, chicken embryo and zebrafish model systems, including high-resolution two-photon microscopy and bioluminescence. We further discuss how imaging technologies are beginning to allow researchers to address the role of regional activation of specific molecular pathways in the metastatic process. These technologies are shedding light, literally, on almost every step of the metastatic process, particularly with regards to the dynamics and plasticity of the disseminating cancer cells and the active participation of the microenvironment in the processes.


Subject(s)
Diagnostic Imaging/methods , Neoplasms/diagnosis , Neoplasms/pathology , Animals , Humans , Neoplasm Metastasis
9.
F1000Res ; 2: 126, 2013.
Article in English | MEDLINE | ID: mdl-24358843

ABSTRACT

UNLABELLED: We have utilized the persistent elevation of fractional excretion (FE) of urate, > 10%, to differentiate cerebral/renal salt wasting (RSW) from the syndrome of inappropriate antidiuretic hormone secretion (SIADH), in which a normalization of FEurate occurs after correction of hyponatremia.  Previous studies suggest as well  that an elevated FEurate with normonatremia, without pre-existing hyponatremia, is also consistent with RSW, including studies demonstrating induction of RSW in rats infused with plasma from normonatremic neurosurgical and Alzheimer's disease patients.  The present studies were designed to test whether precipitates from the urine of normonatremic neurosurgical patients, with either normal or elevated FEurate, and patients with SIADH, display natriuretic activity.   METHODS: Ammonium sulfate precipitates from the urine of 6 RSW and 5 non-RSW Control patients were dialyzed (10 kDa cutoff) to remove the ammonium sulfate, lyophilized, and the reconstituted precipitate was tested for its effect on transcellular transport of (22)Na across LLC-PK1 cells grown to confluency in transwells. RESULTS: Precipitates from 5 of the 6 patients with elevated FEurate and normonatremia significantly inhibited the in vitro transcellular transport of (22)Na above a concentration of 3 µg protein/ml, by 10-25%, versus to vehicle alone, and by 15-40% at concentrations of 5-20 µg/ml as compared to precipitates from 4 of the 5 non-RSW patients with either normal FEurate and normonatremia (2 patients) or with SIADH (2 patients). CONCLUSION: These studies provide further evidence that an elevated FEurate with normonatremia is highly consistent with RSW.  Evidence in the urine of natriuretic activity suggests significant renal excretion of the natriuretic factor. The potentially large source of the natriuretic factor that this could afford, coupled with small analytical sample sizes required by the in-vitro bioassay used here, should facilitate future experimental analysis and allow the natriuretic factor to be investigated as a potential biomarker for RSW.

10.
Cancer Cell ; 21(4): 488-503, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22516258

ABSTRACT

Little is known about the dynamics of cancer cell death in response to therapy in the tumor microenvironment. Intravital microscopy of chemotherapy-treated mouse mammary carcinomas allowed us to follow drug distribution, cell death, and tumor-stroma interactions. We observed associations between vascular leakage and response to doxorubicin, including improved response in matrix metalloproteinase-9 null mice that had increased vascular leakage. Furthermore, we observed CCR2-dependent infiltration of myeloid cells after treatment and that Ccr2 null host mice responded better to treatment with doxorubicin or cisplatin. These data show that the microenvironment contributes critically to drug response via regulation of vascular permeability and innate immune cell infiltration. Thus, live imaging can be used to gain insights into drug responses in situ.


Subject(s)
Drug Resistance, Neoplasm , Mammary Neoplasms, Experimental/drug therapy , Tumor Microenvironment/drug effects , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cisplatin/pharmacokinetics , Cisplatin/therapeutic use , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Macrophages/drug effects , Macrophages/pathology , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/pathology , Receptors, CCR2/genetics , Receptors, CCR2/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL