Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nat Cancer ; 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429414

ABSTRACT

Successful immunotherapy relies on triggering complex responses involving T cell dynamics in tumors and the periphery. Characterizing these responses remains challenging using static human single-cell atlases or mouse models. To address this, we developed a framework for in vivo tracking of tumor-specific CD8+ T cells over time and at single-cell resolution. Our tools facilitate the modeling of gene program dynamics in the tumor microenvironment (TME) and the tumor-draining lymph node (tdLN). Using this approach, we characterize two modes of anti-programmed cell death protein 1 (PD-1) activity, decoupling induced differentiation of tumor-specific activated precursor cells from conventional type 1 dendritic cell (cDC1)-dependent proliferation and recruitment to the TME. We demonstrate that combining anti-PD-1 therapy with anti-4-1BB agonist enhances the recruitment and proliferation of activated precursors, resulting in tumor control. These data suggest that effective response to anti-PD-1 therapy is dependent on sufficient influx of activated precursor CD8+ cells to the TME and highlight the importance of understanding system-level dynamics in optimizing immunotherapies.

2.
Cancer Discov ; 13(12): 2610-2631, 2023 12 12.
Article in English | MEDLINE | ID: mdl-37756565

ABSTRACT

Cancer mortality primarily stems from metastatic recurrence, emphasizing the urgent need for developing effective metastasis-targeted immunotherapies. To better understand the cellular and molecular events shaping metastatic niches, we used a spontaneous breast cancer lung metastasis model to create a single-cell atlas spanning different metastatic stages and regions. We found that premetastatic lungs are infiltrated by inflammatory neutrophils and monocytes, followed by the accumulation of suppressive macrophages with the emergence of metastases. Spatial profiling revealed that metastasis-associated immune cells were present in the metastasis core, with the exception of TREM2+ regulatory macrophages uniquely enriched at the metastatic invasive margin, consistent across both murine models and human patient samples. These regulatory macrophages (Mreg) contribute to the formation of an immune-suppressive niche, cloaking tumor cells from immune surveillance. Our study provides a compendium of immune cell dynamics across metastatic stages and niches, informing the development of metastasis-targeting immunotherapies. SIGNIFICANCE: Temporal and spatial single-cell analysis of metastasis stages revealed new players in modulating immune surveillance and suppression. Our study highlights distinct populations of TREM2 macrophages as modulators of the microenvironment in metastasis, and as the key immune determinant defining metastatic niches, pointing to myeloid checkpoints to improve therapeutic strategies. This article is featured in Selected Articles from This Issue, p. 2489.


Subject(s)
Breast Neoplasms , Lung Neoplasms , Mice , Humans , Animals , Female , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Lung Neoplasms/pathology , Lung/pathology , Macrophages , Tumor Microenvironment , Neoplasm Metastasis/pathology , Membrane Glycoproteins , Receptors, Immunologic
3.
Nat Med ; 29(5): 1191-1200, 2023 05.
Article in English | MEDLINE | ID: mdl-37106166

ABSTRACT

Erythropoietin (Epo) is the master regulator of erythropoiesis and oxygen homeostasis. Despite its physiological importance, the molecular and genomic contexts of the cells responsible for renal Epo production remain unclear, limiting more-effective therapies for anemia. Here, we performed single-cell RNA and transposase-accessible chromatin (ATAC) sequencing of an Epo reporter mouse to molecularly identify Epo-producing cells under hypoxic conditions. Our data indicate that a distinct population of kidney stroma, which we term Norn cells, is the major source of endocrine Epo production in mice. We use these datasets to identify the markers, signaling pathways and transcriptional circuits characteristic of Norn cells. Using single-cell RNA sequencing and RNA in situ hybridization in human kidney tissues, we further provide evidence that this cell population is conserved in humans. These preliminary findings open new avenues to functionally dissect EPO gene regulation in health and disease and may serve as groundwork to improve erythropoiesis-stimulating therapies.


Subject(s)
Anemia , Erythropoietin , Animals , Humans , Mice , Anemia/genetics , Erythropoiesis/genetics , Erythropoietin/genetics , Kidney/metabolism , RNA/metabolism
4.
Int J Rheum Dis ; 26(5): 965-967, 2023 May.
Article in English | MEDLINE | ID: mdl-36599652

ABSTRACT

We report the case of a 65 year old female patient, presenting with a combination of bilateral hearing loss, otalgia, and hyperacusis. Pure tone audiometry revealed mixed bilateral hearing loss. Conventional cranial imaging tests failed to show a significant brain pathology, but fat-suppressed T1-weighted gadolinium-enhanced magnetic resonance imaging scan displayed a diffuse infiltrative skull base process, extending from the nasopharynx to the jugular fossa, and encasing the internal carotid artery. The latter findings, besides elevated inflammatory markers and a positive perinuclear anti-neutrophil cytoplasmic antibody (p-ANCA) led to the diagnosis of ANCA-associated vasculitis. Additional disease manifestations sequentially appeared, including a right peripheral nerve palsy, aortitis, hepatitis, peripheral neuropathy, and uveitis. Therapy with corticosteroids, azathioprine, and then cyclophosphamide brought no evident benefit, but rituximab led to impressive clinical and radiologic improvement.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Antibodies, Antineutrophil Cytoplasmic , Female , Humans , Aged , Hearing Loss, Bilateral , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/diagnosis , Cyclophosphamide/therapeutic use , Rituximab/therapeutic use
6.
Front Cell Infect Microbiol ; 12: 968739, 2022.
Article in English | MEDLINE | ID: mdl-36118038

ABSTRACT

Bladder cancer is the 4th leading cancer in men. Tumor resection followed by bladder instillation of Bacillus Calmette-Guérin (BCG) is the primary treatment for high-risk patients with Non-Muscle Invasive Bladder Cancer (NMIBC) to prevent recurrence and progression to muscle-invasive disease. This treatment, however, lacks efficiency and causes severe adverse effects. Mannose residues are expressed on bladder surfaces and their levels were indicated to be higher in bladder cancer. Intravesical instillations of a recombinant Pseudomonas aeruginosa (PA) overexpressing the mannose-sensitive hemagglutination fimbriae (PA-MSHA), and of a mannose-specific lectin-drug conjugate showed efficiency against NMIBC in murine models of bladder cancer. Urothelial mannosylation facilitates bladder colonization by Uropathogenic E. coli (UPEC) via the interaction with the FimH mannose lectin, positioned at the tip of type 1 fimbria. A recombinant BCG strain overexpressing FimH on its outer surface, exhibited higher attachment and internalization to bladder cancer cells and increased effectivity in treating bladder cancer in mice. Investigating the pattern of mannose expression in NMIBC is important for improving treatment. Here, using tissue microarrays containing multiple normal and cancerous bladder samples, and lectins, we confirm that human bladder cancer cells express high mannose levels. Using UPEC mutants lacking or overexpressing type 1 fimbria, we also demonstrate that tumor-induced hypermannosylation increases type 1 fimbria mediated UPEC attachment to human and mouse bladder cancer. Our results provide an explanation for the effectiveness of PA-MSHA and the FimH-overexpressing BCG and support the hypothesis that mannose-targeted therapy holds potential for improving bladder cancer treatment.


Subject(s)
Mycobacterium bovis , Urinary Bladder Neoplasms , Uropathogenic Escherichia coli , Animals , BCG Vaccine , Fimbriae Proteins/metabolism , Humans , Lectins , Mannose , Mannose-Binding Lectins , Mice , Pseudomonas aeruginosa/metabolism , Urinary Bladder Neoplasms/pathology , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/metabolism
7.
Cell ; 185(8): 1373-1388.e20, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35381199

ABSTRACT

Systemic sclerosis (scleroderma, SSc) is an incurable autoimmune disease with high morbidity and mortality rates. Here, we conducted a population-scale single-cell genomic analysis of skin and blood samples of 56 healthy controls and 97 SSc patients at different stages of the disease. We found immune compartment dysfunction only in a specific subtype of diffuse SSc patients but global dysregulation of the stromal compartment, particularly in a previously undefined subset of LGR5+-scleroderma-associated fibroblasts (ScAFs). ScAFs are perturbed morphologically and molecularly in SSc patients. Single-cell multiome profiling of stromal cells revealed ScAF-specific markers, pathways, regulatory elements, and transcription factors underlining disease development. Systematic analysis of these molecular features with clinical metadata associates specific ScAF targets with disease pathogenesis and SSc clinical traits. Our high-resolution atlas of the sclerodermatous skin spectrum will enable a paradigm shift in the understanding of SSc disease and facilitate the development of biomarkers and therapeutic strategies.


Subject(s)
Scleroderma, Systemic , Cells, Cultured , Fibroblasts/metabolism , Fibrosis , Humans , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/genetics , Skin/metabolism
8.
APMIS ; 130(5): 270-275, 2022 May.
Article in English | MEDLINE | ID: mdl-35218080

ABSTRACT

We report a case of Staphylococcus warneri native valve endocarditis in an immunocompetent healthy adult, without known risk factors for infective endocarditis, two months following COVID-19 infection, who recovered with conservative treatment. Additionally, we reviewed previous cases of native valve endocarditis caused by Staphylococcus warneri and summarized the main clinical implications.


Subject(s)
COVID-19 , Endocarditis, Bacterial , Endocarditis , Staphylococcal Infections , Adult , Aortic Valve , Endocarditis, Bacterial/diagnosis , Endocarditis, Bacterial/drug therapy , Humans , Staphylococcal Infections/drug therapy , Staphylococcus
11.
Nat Med ; 27(6): 1043-1054, 2021 06.
Article in English | MEDLINE | ID: mdl-34017133

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are prevalent liver conditions that underlie the development of life-threatening cirrhosis, liver failure and liver cancer. Chronic necro-inflammation is a critical factor in development of NASH, yet the cellular and molecular mechanisms of immune dysregulation in this disease are poorly understood. Here, using single-cell transcriptomic analysis, we comprehensively profiled the immune composition of the mouse liver during NASH. We identified a significant pathology-associated increase in hepatic conventional dendritic cells (cDCs) and further defined their source as NASH-induced boost in cycling of cDC progenitors in the bone marrow. Analysis of blood and liver from patients on the NAFLD/NASH spectrum showed that type 1 cDCs (cDC1) were more abundant and activated in disease. Sequencing of physically interacting cDC-T cell pairs from liver-draining lymph nodes revealed that cDCs in NASH promote inflammatory T cell reprogramming, previously associated with NASH worsening. Finally, depletion of cDC1 in XCR1DTA mice or using anti-XCL1-blocking antibody attenuated liver pathology in NASH mouse models. Overall, our study provides a comprehensive characterization of cDC biology in NASH and identifies XCR1+ cDC1 as an important driver of liver pathology.


Subject(s)
Dendritic Cells/immunology , Fatty Liver/immunology , Non-alcoholic Fatty Liver Disease/immunology , Receptors, Chemokine/genetics , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Dendritic Cells/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/pathology , Female , Humans , Liver/immunology , Liver/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Chemokine/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
Nat Med ; 27(3): 491-503, 2021 03.
Article in English | MEDLINE | ID: mdl-33619369

ABSTRACT

Multiple myeloma (MM) is a neoplastic plasma-cell disorder characterized by clonal proliferation of malignant plasma cells. Despite extensive research, disease heterogeneity within and between treatment-resistant patients is poorly characterized. In the present study, we conduct a prospective, multicenter, single-arm clinical trial (NCT04065789), combined with longitudinal single-cell RNA-sequencing (scRNA-seq) to study the molecular dynamics of MM resistance mechanisms. Newly diagnosed MM patients (41), who either failed to respond or experienced early relapse after a bortezomib-containing induction regimen, were enrolled to evaluate the safety and efficacy of a daratumumab, carfilzomib, lenalidomide and dexamethasone combination. The primary clinical endpoint was safety and tolerability. Secondary endpoints included overall response rate, progression-free survival and overall survival. Treatment was safe and well tolerated; deep and durable responses were achieved. In prespecified exploratory analyses, comparison of 41 primary refractory and early relapsed patients, with 11 healthy subjects and 15 newly diagnosed MM patients, revealed new MM molecular pathways of resistance, including hypoxia tolerance, protein folding and mitochondria respiration, which generalized to larger clinical cohorts (CoMMpass). We found peptidylprolyl isomerase A (PPIA), a central enzyme in the protein-folding response pathway, as a potential new target for resistant MM. CRISPR-Cas9 deletion of PPIA or inhibition of PPIA with a small molecule inhibitor (ciclosporin) significantly sensitizes MM tumor cells to proteasome inhibitors. Together, our study defines a roadmap for integrating scRNA-seq in clinical trials, identifies a signature of highly resistant MM patients and discovers PPIA as a potent therapeutic target for these tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Single-Cell Analysis/methods , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Case-Control Studies , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Lenalidomide/administration & dosage , Male , Middle Aged , Neoplasm Recurrence, Local , Oligopeptides/administration & dosage , Treatment Outcome
13.
Oncoimmunology ; 8(6): e1581531, 2019.
Article in English | MEDLINE | ID: mdl-31069151

ABSTRACT

Fusobacterium nucleatum (F. nucleatum) is an oral anaerobe found to be enriched in colorectal cancer (CRC). Presence of F. nucleatum in CRC has been correlated with resistance to chemotherapy and poor prognosis. We previously demonstrated that the Fap2 outer-surface protein of F. nucleatum binds and activates the human inhibitory receptor TIGIT which is expressed by T and Natural Killer (NK) cells, and inhibits anti-tumor immunity. Here we show that F. nucleatum also binds and activates the human inhibitory receptor CEACAM1 leading to inhibition of T and NK cells activities. Our results suggest that using CEACAM1 and TIGIT inhibitors and specific targeting of fusobacteria should be considered for treating fusobacteria-colonized tumors.

14.
Oncoimmunology ; 8(4): e1553487, 2019.
Article in English | MEDLINE | ID: mdl-30906650

ABSTRACT

We previously showed that the colorectal cancer colonizing bacterium Fusobacterium nucleatum protects tumors from immune cell attack via binding of the fusbacterial Fap2 outer-membrane protein to TIGIT, a checkpoint inhibitory receptor expressed on T cells and NK cells. Helicobacter pylori, the causative agent for peptic ulcer disease, is associated with the development of gastric adenocarcinoma and MALT lymphoma. The HopQ outer-membrane adhesin of H. pylori was recently shown to bind carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) including CEACAM1, an inhibitory receptor expressed mainly by activated T and NK cells. Here we investigated the possibility that similar to Fap2, HopQ can also inhibit immune cell activities by interacting with CEACAM1. We used several approaches to confirm that HopQ indeed interacts with CEACAM1, and show that CEACAM1-mediated activation by HopQ, may inhibit NK and T cell functions.

15.
Sci Rep ; 9(1): 1351, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718650

ABSTRACT

Rheumatoid Arthritis (RA) causes chronic inflammation of joints. The cytokines TNFα and IFNγ are central players in RA, however their source has not been fully elucidated. Natural Killer (NK) cells are best known for their role in elimination of viral-infected and transformed cells, and they secrete pro-inflammatory cytokines. NK cells are present in the synovial fluids (SFs) of RA patients and are considered to be important in bone destruction. However, the phenotype and function of NK cells in the SFs of patients with erosive deformative RA (DRA) versus non-deformative RA (NDRA) is poorly characterized. Here we characterize the NK cell populations present in the blood and SFs of DRA and NDRA patients. We demonstrate that a distinct population of activated synovial fluid NK (sfNK) cells constitutes a large proportion of immune cells found in the SFs of DRA patients. We discovered that although sfNK cells in both DRA and NDRA patients have similar phenotypes, they function differently. The DRA sfNK secrete more TNFα and IFNγ upon exposure to IL-2 and IL-15. Consequently, we suggest that sfNK cells may be a marker for more severely destructive RA disease.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Killer Cells, Natural/immunology , Synovial Fluid/immunology , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/blood , Case-Control Studies , Female , Humans , Interferon-gamma/metabolism , Male , Middle Aged , Phenotype , Receptors, Natural Killer Cell/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Immunity ; 48(5): 951-962.e5, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768178

ABSTRACT

Natural killer cells (NKs) are abundant in the human decidua, regulating trophoblast invasion and angiogenesis. Several diseases of poor placental development are associated with first pregnancies, so we thus looked to characterize differences in decidual NKs (dNKs) in first versus repeated pregnancies. We discovered a population found in repeated pregnancies, which has a unique transcriptome and epigenetic signature, and is characterized by high expression of the receptors NKG2C and LILRB1. We named these cells Pregnancy Trained decidual NK cells (PTdNKs). PTdNKs have open chromatin around the enhancers of IFNG and VEGFA. Activation of PTdNKs led to increased production and secretion of IFN-γ and VEGFα, with the latter supporting vascular sprouting and tumor growth. The precursors of PTdNKs seem to be found in the endometrium. Because repeated pregnancies are associated with improved placentation, we propose that PTdNKs, which are present primarily in repeated pregnancies, might be involved in proper placentation.


Subject(s)
Immunologic Memory/immunology , Killer Cells, Natural/immunology , Transcriptome/immunology , Uterus/immunology , Animals , Cell Line, Tumor , Decidua/immunology , Decidua/metabolism , Female , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Pregnancy , Uterus/cytology , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
17.
Cell Rep ; 20(1): 40-47, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28683322

ABSTRACT

Urinary tract infection (UTI) is the most common type of bacterial infection in humans. Fifty percent of all women will experience at least one UTI in their lifetime, with uropathogenic Escherichia coli (UPEC) accounting for 80% of reported cases. UTI evokes a complex, well-timed immune response that is crucial for bacterial clearance. The majority of immune cells participating in the immune response are absent from the healthy bladder, and the mechanisms used to recruit them upon UTI are not fully understood. Here, we show that immediately after UPEC infection, bladder epithelial cells secrete stromal cell-derived factor 1 (SDF-1), initiating immune cell accumulation at the site of infection. SDF-1 blockade significantly reduced immune cell migration to the infected bladder, resulting in severe exacerbation of infection. We also show that FimH, the adhesin of type 1 fimbria, one of UPEC's virulence factors, is directly involved in the secretion of SDF-1 upon UTI.


Subject(s)
Chemokine CXCL12/immunology , Immunity, Innate , Urinary Tract Infections/immunology , Adhesins, Escherichia coli/immunology , Animals , Chemokine CXCL12/metabolism , Enteropathogenic Escherichia coli/immunology , Enteropathogenic Escherichia coli/pathogenicity , Female , Fimbriae Proteins/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , T-Lymphocytes/immunology , Urinary Tract Infections/microbiology , Urothelium/immunology , Urothelium/metabolism
18.
Cell Host Microbe ; 20(4): 527-534, 2016 Oct 12.
Article in English | MEDLINE | ID: mdl-27736647

ABSTRACT

Natural killer (NK) cells form an important arm of the innate immune system and function to combat a wide range of invading pathogens, ranging from viruses to bacteria. However, the means by which NK cells accomplish recognition of pathogens with a limited repertoire of receptors remain largely unknown. In the current study, we describe the recognition of an emerging fungal pathogen, Candida glabrata, by the human NK cytotoxic receptor NKp46 and its mouse ortholog, NCR1. Using NCR1 knockout mice, we observed that this receptor-mediated recognition was crucial for controlling C. glabrata infection in vitro and in vivo. Finally, we delineated the fungal ligands to be the C. glabrata adhesins Epa1, Epa6, and Epa7 and demonstrated that clearance of systemic C. glabrata infections in vivo depends on their recognition by NCR1. As NKp46 and NCR1 have been previously shown to bind viral adhesion receptors, we speculate that NKp46/NCR1 may be a novel type of pattern recognition receptor.


Subject(s)
Antigens, Ly/metabolism , Candida glabrata/immunology , Fungal Proteins/metabolism , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Animals , Antigens, Ly/genetics , Candidiasis/immunology , Disease Models, Animal , Humans , Mice, Inbred BALB C , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/genetics
19.
Oncoimmunology ; 4(9): e1038690, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26405611

ABSTRACT

Fusobacterium nucleatum is present in colon cancers where it was shown to generate a proinflammatory microenvironment that supports colorectal neoplasia progression. Remarkably, alongside with proinflammatory stimulation, fusobacteria also inhibit cytotoxicity of immune cells. Thus, it appears as if tumors exploit fusobacteria to generate a favorable proinflammatory and anti-cytotoxic microenvironment.

20.
Immunity ; 42(2): 344-355, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680274

ABSTRACT

Bacteria, such as Fusobacterium nucleatum, are present in the tumor microenvironment. However, the immunological consequences of intra-tumoral bacteria remain unclear. Here, we have shown that natural killer (NK) cell killing of various tumors is inhibited in the presence of various F. nucleatum strains. Our data support that this F. nucleatum-mediated inhibition is mediated by human, but not by mouse TIGIT, an inhibitory receptor present on all human NK cells and on various T cells. Using a library of F. nucleatum mutants, we found that the Fap2 protein of F. nucleatum directly interacted with TIGIT, leading to the inhibition of NK cell cytotoxicity. We have further demonstrated that tumor-infiltrating lymphocytes expressed TIGIT and that T cell activities were also inhibited by F. nucleatum via Fap2. Our results identify a bacterium-dependent, tumor-immune evasion mechanism in which tumors exploit the Fap2 protein of F. nucleatum to inhibit immune cell activity via TIGIT.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/microbiology , Colonic Neoplasms/immunology , Colonic Neoplasms/microbiology , Fusobacterium nucleatum/immunology , Receptors, Immunologic/immunology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , Bacterial Outer Membrane Proteins/immunology , Cell Line , Cell Proliferation , Humans , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...