Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Psychiatry ; 26(7): 2837-2853, 2021 07.
Article in English | MEDLINE | ID: mdl-33767348

ABSTRACT

The high comorbidity between obesity and mental disorders, such as depression and anxiety, often exacerbates metabolic and neurological symptoms significantly. However, neural mechanisms that underlie reciprocal control of feeding and mental states are largely elusive. Here we report that melanocortin 4 receptor (MC4R) neurons located in the dorsal bed nucleus of the stria terminus (dBNST) engage in the regulation of mentally associated weight gain by receiving GABAergic projections from hypothalamic AgRP neurons onto α5-containing GABAA receptors and serotonergic afferents onto 5-HT3 receptors. Chronic treatment with a high-fat diet (HFD) significantly blunts the hyperexcitability of AgRP neurons in response to not only hunger but also anxiety and depression-like stimuli. Such HFD-mediated desensitization reduces GABAergic outputs from AgRP neurons to downstream MC4RdBNST neurons, resulting in severe mental dysregulation. Genetic enhancement of the GABAAR-α5 or suppression of the 5-HT3R within the MC4RdBNST neurons not only abolishes HFD-induced anxiety and depression but also robustly reduces body weight by suppression of food intake. To gain further translational insights, we revealed that combined treatment of zonisamide (enhancing the GABAAR-α5 signaling) and granisetron (a selective 5-HT3R antagonist) alleviates mental dysfunction and yields a robust reversal of diet-induced obesity by reducing total calorie intake and altering food preference towards a healthy low-fat diet. Our results unveil a neural mechanism for reciprocal control of appetite and mental states, which culminates in a novel zonisamide-granisetron cocktail therapy for potential tackling the psychosis-obesity comorbidity.


Subject(s)
Depressive Disorder , Serotonin , Agouti-Related Protein , Anxiety , Depression , Humans , Obesity , gamma-Aminobutyric Acid
2.
Genet Med ; 22(2): 301-308, 2020 02.
Article in English | MEDLINE | ID: mdl-31467446

ABSTRACT

PURPOSE: Fetal fraction (FF) is the percent of cell-free DNA (cfDNA) in the mother's peripheral blood that is of fetal origin, which plays a pivotal role in noninvasive prenatal screening (NIPS). We present a method that can reliably estimate FFs by examining autosome single-nucleotide polymorphisms (SNPs). METHODS: Even at a very low sequencing depth, there are plenty of SNPs covered by more than one read. At those SNPs, we define read heterozygosity and demonstrate that the percent of read heterozygosity is a function of FF, which allows FF to be inferred. RESULTS: We first demonstrated the effectiveness of our method in inferring FF. Then we used the inferred FF as an informative alternative prior to computing Bayes factors to test for aneuploidy, and observed better power than the Z-test. In analysis of clinical samples, we were able to identify female-male twins thanks to the accurate FF inference. CONCLUSION: Knowing FF improves efficacy of NIPS. It brings a powerful Bayesian method, allows "no call" for samples with small FFs, renders screening for XXY syndrome simpler, and permits an adaptive design to sequence at a higher depth for samples with small FFs.


Subject(s)
Cell-Free Nucleic Acids/analysis , Fetal Development/genetics , Noninvasive Prenatal Testing/methods , Chromosome Aberrations , Female , Fetus , High-Throughput Nucleotide Sequencing/methods , Humans , Polymorphism, Single Nucleotide/genetics , Pregnancy , Prenatal Care , Prenatal Diagnosis/methods , Sequence Analysis, DNA/methods
3.
Genet Med ; 22(2): 450, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31822850

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Proc Natl Acad Sci U S A ; 114(4): 770-775, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28074040

ABSTRACT

Hepatitis A virus (HAV) infects ∼1.4 million people annually and, although there is a vaccine, there are no licensed therapeutic drugs. HAV is unusually stable (making disinfection problematic) and little is known of how it enters cells and releases its RNA. Here we report a potent HAV-specific monoclonal antibody, R10, which neutralizes HAV infection by blocking attachment to the host cell. High-resolution cryo-EM structures of HAV full and empty particles and of the complex of HAV with R10 Fab reveal the atomic details of antibody binding and point to a receptor recognition site at the pentamer interface. These results, together with our observation that the R10 Fab destabilizes the capsid, suggest the use of a receptor mimic mechanism to neutralize virus infection, providing new opportunities for therapeutic intervention.


Subject(s)
Antibodies, Neutralizing/immunology , Hepatitis A virus/immunology , Animals , Antibodies, Monoclonal/immunology , Binding Sites/immunology , Capsid/immunology , Capsid Proteins/immunology , Female , Humans , Immunoglobulin Fab Fragments/immunology , Mice , Mice, Inbred BALB C
5.
Neuro Oncol ; 26(5): 826-839, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38237157

ABSTRACT

BACKGROUND: Glioblastomas (GBMs) are central nervous system tumors that resist standard-of-care interventions and even immune checkpoint blockade. Myeloid cells in the tumor microenvironment can contribute to GBM progression; therefore, emerging immunotherapeutic approaches include reprogramming these cells to achieve desirable antitumor activity. Triggering receptor expressed on myeloid cells 2 (TREM2) is a myeloid signaling regulator that has been implicated in a variety of cancers and neurological diseases with contrasting functions, but its role in GBM immunopathology and progression is still under investigation. METHODS: Our reverse translational investigations leveraged single-cell RNA sequencing and cytometry of human gliomas to characterize TREM2 expression across myeloid subpopulations. Using 2 distinct murine glioma models, we examined the role of Trem2 on tumor progression and immune modulation of myeloid cells. Furthermore, we designed a method of tracking phagocytosis of glioma cells in vivo and employed in vitro assays to mechanistically understand the influence of TREM2 signaling on tumor uptake. RESULTS: We discovered that TREM2 expression does not correlate with immunosuppressive pathways, but rather showed strong a positive association with the canonical phagocytosis markers lysozyme (LYZ) and macrophage scavenger receptor (CD163) in gliomas. While Trem2 deficiency was found to be dispensable for gliomagenesis, Trem2+ myeloid cells display enhanced tumor uptake compared to Trem2- cells. Mechanistically, we demonstrate that TREM2 mediates phagocytosis via Syk signaling. CONCLUSIONS: These results indicate that TREM2 is not associated with immunosuppression in gliomas. Instead, TREM2 is an important regulator of phagocytosis that may be exploited as a potential therapeutic strategy for brain tumors.


Subject(s)
Brain Neoplasms , Glioblastoma , Membrane Glycoproteins , Phagocytosis , Receptors, Immunologic , Animals , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice , Humans , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Tumor Microenvironment , Myeloid Cells/metabolism , Mice, Inbred C57BL , Tumor Cells, Cultured , Signal Transduction
6.
bioRxiv ; 2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37066184

ABSTRACT

Glioblastomas (GBMs) are tumors of the central nervous system that remain recalcitrant to both standard of care chemo-radiation and immunotherapies. Emerging approaches to treat GBMs include depletion or re-education of innate immune cells including microglia (MG) and macrophages (MACs). Here we show myeloid cell restricted expression of triggering receptor expressed on myeloid cells 2 (TREM2) across low- and high-grade human gliomas. TREM2 expression did not correlate with immunosuppressive pathways, but rather showed strong positive association with phagocytosis markers such as lysozyme (LYZ) and CD163 in gliomas. In line with these observations in patient tumors, Trem2-/- mice did not exhibit improved survival compared to wildtype (WT) mice when implanted with mouse glioma cell lines, unlike observations previously seen in peripheral tumor models. Gene expression profiling revealed pathways related to inflammation, adaptive immunity, and autophagy that were significantly downregulated in tumors from Trem2-/- mice compared to WT tumors. Using ZsGreen-expressing CT-2A orthotopic implants, we found higher tumor antigen engulfment in Trem2+ MACs, MG, and dendritic cells. Our data uncover TREM2 as an important immunomodulator in gliomas and inducing TREM2 mediated phagocytosis can be a potential immunotherapeutic strategy for brain tumors.

7.
Cancer Cell ; 41(7): 1363-1380.e7, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37327788

ABSTRACT

Inactivating STK11/LKB1 mutations are genomic drivers of primary resistance to immunotherapy in KRAS-mutated lung adenocarcinoma (LUAD), although the underlying mechanisms remain unelucidated. We find that LKB1 loss results in enhanced lactate production and secretion via the MCT4 transporter. Single-cell RNA profiling of murine models indicates that LKB1-deficient tumors have increased M2 macrophage polarization and hypofunctional T cells, effects that could be recapitulated by the addition of exogenous lactate and abrogated by MCT4 knockdown or therapeutic blockade of the lactate receptor GPR81 expressed on immune cells. Furthermore, MCT4 knockout reverses the resistance to PD-1 blockade induced by LKB1 loss in syngeneic murine models. Finally, tumors from STK11/LKB1 mutant LUAD patients demonstrate a similar phenotype of enhanced M2-macrophages polarization and hypofunctional T cells. These data provide evidence that lactate suppresses antitumor immunity and therapeutic targeting of this pathway is a promising strategy to reversing immunotherapy resistance in STK11/LKB1 mutant LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Animals , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/therapy , Adenocarcinoma of Lung/metabolism , Lactates/metabolism , Lactates/pharmacology , Lactates/therapeutic use , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Macrophages , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
8.
Cancer Cell ; 41(6): 1032-1047.e4, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37311413

ABSTRACT

Multiple myeloma remains an incurable disease, and the cellular and molecular evolution from precursor conditions, including monoclonal gammopathy of undetermined significance and smoldering multiple myeloma, is incompletely understood. Here, we combine single-cell RNA and B cell receptor sequencing from fifty-two patients with myeloma precursors in comparison with myeloma and normal donors. Our comprehensive analysis reveals early genomic drivers of malignant transformation, distinct transcriptional features, and divergent clonal expansion in hyperdiploid versus non-hyperdiploid samples. Additionally, we observe intra-patient heterogeneity with potential therapeutic implications and identify distinct patterns of evolution from myeloma precursor disease to myeloma. We also demonstrate distinctive characteristics of the microenvironment associated with specific genomic changes in myeloma cells. These findings add to our knowledge about myeloma precursor disease progression, providing valuable insights into patient risk stratification, biomarker discovery, and possible clinical applications.


Subject(s)
Biomedical Research , Multiple Myeloma , Smoldering Multiple Myeloma , Humans , Multiple Myeloma/genetics , Aneuploidy , Disease Progression , Tumor Microenvironment/genetics
9.
Cancer Cell ; 41(8): 1407-1426.e9, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37419119

ABSTRACT

Understanding tumor microenvironment (TME) reprogramming in gastric adenocarcinoma (GAC) progression may uncover novel therapeutic targets. Here, we performed single-cell profiling of precancerous lesions, localized and metastatic GACs, identifying alterations in TME cell states and compositions as GAC progresses. Abundant IgA+ plasma cells exist in the premalignant microenvironment, whereas immunosuppressive myeloid and stromal subsets dominate late-stage GACs. We identified six TME ecotypes (EC1-6). EC1 is exclusive to blood, while EC4, EC5, and EC2 are highly enriched in uninvolved tissues, premalignant lesions, and metastases, respectively. EC3 and EC6, two distinct ecotypes in primary GACs, associate with histopathological and genomic characteristics, and survival outcomes. Extensive stromal remodeling occurs in GAC progression. High SDC2 expression in cancer-associated fibroblasts (CAFs) is linked to aggressive phenotypes and poor survival, and SDC2 overexpression in CAFs contributes to tumor growth. Our study provides a high-resolution GAC TME atlas and underscores potential targets for further investigation.


Subject(s)
Adenocarcinoma , Cancer-Associated Fibroblasts , Precancerous Conditions , Stomach Neoplasms , Humans , Ecotype , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Adenocarcinoma/pathology , Cancer-Associated Fibroblasts/pathology , Precancerous Conditions/pathology , Stromal Cells/pathology , Tumor Microenvironment
10.
Nat Med ; 29(6): 1550-1562, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37248301

ABSTRACT

Tumor-infiltrating T cells offer a promising avenue for cancer treatment, yet their states remain to be fully characterized. Here we present a single-cell atlas of T cells from 308,048 transcriptomes across 16 cancer types, uncovering previously undescribed T cell states and heterogeneous subpopulations of follicular helper, regulatory and proliferative T cells. We identified a unique stress response state, TSTR, characterized by heat shock gene expression. TSTR cells are detectable in situ in the tumor microenvironment across various cancer types, mostly within lymphocyte aggregates or potential tertiary lymphoid structures in tumor beds or surrounding tumor edges. T cell states/compositions correlated with genomic, pathological and clinical features in 375 patients from 23 cohorts, including 171 patients who received immune checkpoint blockade therapy. We also found significantly upregulated heat shock gene expression in intratumoral CD4/CD8+ cells following immune checkpoint blockade treatment, particularly in nonresponsive tumors, suggesting a potential role of TSTR cells in immunotherapy resistance. Our well-annotated T cell reference maps, web portal and automatic alignment/annotation tool could provide valuable resources for T cell therapy optimization and biomarker discovery.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/metabolism , Immunotherapy , Tumor Microenvironment
11.
Blood Cancer Discov ; 3(5): 428-443, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35687817

ABSTRACT

Follicular lymphoma (FL) is a B-cell malignancy with a complex tumor microenvironment that is rich in nonmalignant immune cells. We applied single-cell RNA sequencing to characterize the diverse tumor and immune cell populations of FL and identified major phenotypic subsets of FL T cells, including a cytotoxic CD4 T-cell population. We characterized four major FL subtypes with differential representation or relative depletion of distinct T-cell subsets. By integrating exome sequencing, we observed that somatic mutations are associated with, but not definitive for, reduced MHC expression on FL cells. In turn, expression of MHCII genes by FL cells was associated with significant differences in the proportions and targetable immunophenotypic characteristics of T cells. This provides a classification framework of the FL microenvironment in association with FL genotypes and MHC expression, and informs different potential immunotherapeutic strategies based upon tumor cell MHCII expression. SIGNIFICANCE: We have characterized the FL-infiltrating T cells, identified cytotoxic CD4 T cells as an important component that is associated with tumor cell-intrinsic characteristics, and identified sets of targetable immune checkpoints on T cells that differed from FLs with normal versus low MHC expression. See related commentary by Melnick, p. 374. This article is highlighted in the In This Issue feature, p. 369.


Subject(s)
Lymphoma, Follicular , Humans , Immunophenotyping , Lymphoma, Follicular/genetics , Mutation , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/genetics
12.
Cancer Discov ; 12(11): 2626-2645, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36098652

ABSTRACT

Tumor-infiltrating B and plasma cells (TIB) are prevalent in lung adenocarcinoma (LUAD); however, they are poorly characterized. We performed paired single-cell RNA and B-cell receptor (BCR) sequencing of 16 early-stage LUADs and 47 matching multiregion normal tissues. By integrative analysis of ∼50,000 TIBs, we define 12 TIB subsets in the LUAD and adjacent normal ecosystems and demonstrate extensive remodeling of TIBs in LUADs. Memory B cells and plasma cells (PC) were highly enriched in tumor tissues with more differentiated states and increased frequencies of somatic hypermutation. Smokers exhibited markedly elevated PCs and PCs with distinct differentiation trajectories. BCR clonotype diversity increased but clonality decreased in LUADs, smokers, and with increasing pathologic stage. TIBs were mostly localized within CXCL13+ lymphoid aggregates, and immune cell sources of CXCL13 production evolved with LUAD progression and included elevated fractions of CD4 regulatory T cells. This study provides a spatial landscape of TIBs in early-stage LUAD. SIGNIFICANCE: While TIBs are highly enriched in LUADs, they are poorly characterized. This study provides a much-needed understanding of the transcriptional, clonotypic states and phenotypes of TIBs, unraveling their potential roles in the immunopathology of early-stage LUADs and constituting a road map for the development of TIB-targeted immunotherapies for the treatment of this morbid malignancy. This article is highlighted in the In This Issue feature, p. 2483.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Humans , Plasma Cells/pathology , Ecosystem , Lung Neoplasms/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma/genetics , Prognosis
13.
Nat Med ; 27(1): 141-151, 2021 01.
Article in English | MEDLINE | ID: mdl-33398161

ABSTRACT

Intratumoral heterogeneity (ITH) is a fundamental property of cancer; however, the origins of ITH remain poorly understood. We performed single-cell transcriptome profiling of peritoneal carcinomatosis (PC) from 15 patients with gastric adenocarcinoma (GAC), constructed a map of 45,048 PC cells, profiled the transcriptome states of tumor cell populations, incisively explored ITH of malignant PC cells and identified significant correlates with patient survival. The links between tumor cell lineage/state compositions and ITH were illustrated at transcriptomic, genotypic, molecular and phenotypic levels. We uncovered the diversity in tumor cell lineage/state compositions in PC specimens and defined it as a key contributor to ITH. Single-cell analysis of ITH classified PC specimens into two subtypes that were prognostically independent of clinical variables, and a 12-gene prognostic signature was derived and validated in multiple large-scale GAC cohorts. The prognostic signature appears fundamental to GAC carcinogenesis and progression and could be practical for patient stratification.


Subject(s)
Adenocarcinoma/secondary , Stomach Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Cell Lineage/genetics , Chromosomes, Human, Pair 17/genetics , Cohort Studies , DNA Copy Number Variations , Female , Gene Expression Profiling , Genetic Variation , Humans , Male , Middle Aged , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Prognosis , RNA-Seq , Single-Cell Analysis , Stomach Neoplasms/genetics
14.
Nat Commun ; 12(1): 5606, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556668

ABSTRACT

Immune checkpoint therapy (ICT) provides substantial clinical benefits to cancer patients, but a large proportion of cancers do not respond to ICT. To date, the genomic underpinnings of primary resistance to ICT remain elusive. Here, we performed immunogenomic analysis of data from TCGA and clinical trials of anti-PD-1/PD-L1 therapy, with a particular focus on homozygous deletion of 9p21.3 (9p21 loss), one of the most frequent genomic defects occurring in ~13% of all cancers. We demonstrate that 9p21 loss confers "cold" tumor-immune phenotypes, characterized by reduced abundance of tumor-infiltrating leukocytes (TILs), particularly, T/B/NK cells, altered spatial TILs patterns, diminished immune cell trafficking/activation, decreased rate of PD-L1 positivity, along with activation of immunosuppressive signaling. Notably, patients with 9p21 loss exhibited significantly lower response rates to ICT and worse outcomes, which were corroborated in eight ICT trials of >1,000 patients. Further, 9p21 loss synergizes with PD-L1/TMB for patient stratification. A "response score" was derived by incorporating 9p21 loss, PD-L1 expression and TMB levels in pre-treatment tumors, which outperforms PD-L1, TMB, and their combination in identifying patients with high likelihood of achieving sustained response from otherwise non-responders. Moreover, we describe potential druggable targets in 9p21-loss tumors, which could be exploited to design rational therapeutic interventions.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Drug Resistance, Neoplasm/immunology , Immune Checkpoint Inhibitors/therapeutic use , Tumor Microenvironment/immunology , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Drug Resistance, Neoplasm/genetics , Homozygote , Humans , Immune Tolerance , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/mortality , Prognosis , Signal Transduction/immunology
15.
Nat Commun ; 12(1): 2877, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001881

ABSTRACT

The mechanisms driving therapeutic resistance and poor outcomes of mantle cell lymphoma (MCL) are incompletely understood. We characterize the cellular and molecular heterogeneity within and across patients and delineate the dynamic evolution of tumor and immune cell compartments at single cell resolution in longitudinal specimens from ibrutinib-sensitive patients and non-responders. Temporal activation of multiple cancer hallmark pathways and acquisition of 17q are observed in a refractory MCL. Multi-platform validation is performed at genomic and cellular levels in PDX models and larger patient cohorts. We demonstrate that due to 17q gain, BIRC5/survivin expression is upregulated in resistant MCL tumor cells and targeting BIRC5 results in marked tumor inhibition in preclinical models. In addition, we discover notable differences in the tumor microenvironment including progressive dampening of CD8+ T cells and aberrant cell-to-cell communication networks in refractory MCLs. This study reveals diverse and dynamic tumor and immune programs underlying therapy resistance in MCL.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Profiling/methods , Genetic Heterogeneity , Lymphoma, Mantle-Cell/genetics , Single-Cell Analysis/methods , Tumor Microenvironment/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Humans , Imidazoles/pharmacology , Lymphoma, Mantle-Cell/diagnostic imaging , Lymphoma, Mantle-Cell/drug therapy , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Naphthoquinones/pharmacology , Positron Emission Tomography Computed Tomography/methods , Sequence Analysis, RNA/methods , Xenograft Model Antitumor Assays/methods
16.
Science ; 374(6575): 1632-1640, 2021 Dec 24.
Article in English | MEDLINE | ID: mdl-34941392

ABSTRACT

Gut bacteria modulate the response to immune checkpoint blockade (ICB) treatment in cancer, but the effect of diet and supplements on this interaction is not well studied. We assessed fecal microbiota profiles, dietary habits, and commercially available probiotic supplement use in melanoma patients and performed parallel preclinical studies. Higher dietary fiber was associated with significantly improved progression-free survival in 128 patients on ICB, with the most pronounced benefit observed in patients with sufficient dietary fiber intake and no probiotic use. Findings were recapitulated in preclinical models, which demonstrated impaired treatment response to anti­programmed cell death 1 (anti­PD-1)­based therapy in mice receiving a low-fiber diet or probiotics, with a lower frequency of interferon-γ­positive cytotoxic T cells in the tumor microenvironment. Together, these data have clinical implications for patients receiving ICB for cancer.


Subject(s)
Dietary Fiber , Gastrointestinal Microbiome , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/therapy , Probiotics , Animals , Cohort Studies , Fatty Acids, Volatile/analysis , Fecal Microbiota Transplantation , Feces/chemistry , Feces/microbiology , Female , Humans , Immunotherapy , Male , Melanoma/immunology , Melanoma/microbiology , Melanoma, Experimental/immunology , Melanoma, Experimental/microbiology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Progression-Free Survival , T-Lymphocytes
17.
Cancer Discov ; 11(10): 2506-2523, 2021 10.
Article in English | MEDLINE | ID: mdl-33972311

ABSTRACT

Little is known of the geospatial architecture of individual cell populations in lung adenocarcinoma (LUAD) evolution. Here, we perform single-cell RNA sequencing of 186,916 cells from five early-stage LUADs and 14 multiregion normal lung tissues of defined spatial proximities from the tumors. We show that cellular lineages, states, and transcriptomic features geospatially evolve across normal regions to LUADs. LUADs also exhibit pronounced intratumor cell heterogeneity within single sites and transcriptional lineage-plasticity programs. T regulatory cell phenotypes are increased in normal tissues with proximity to LUAD, in contrast to diminished signatures and fractions of cytotoxic CD8+ T cells, antigen-presenting macrophages, and inflammatory dendritic cells. We further find that the LUAD ligand-receptor interactome harbors increased expression of epithelial CD24, which mediates protumor phenotypes. These data provide a spatial atlas of LUAD evolution, and a resource for identification of targets for its treatment. SIGNIFICANCE: The geospatial ecosystem of the peripheral lung and early-stage LUAD is not known. Our multiregion single-cell sequencing analyses unravel cell populations, states, and phenotypes in the spatial and ecologic evolution of LUAD from the lung that comprise high-potential targets for early interception.This article is highlighted in the In This Issue feature, p. 2355.


Subject(s)
Adenocarcinoma of Lung/pathology , CD8-Positive T-Lymphocytes , Lung Neoplasms/pathology , Tumor Microenvironment , Humans , Single-Cell Analysis
18.
Nat Med ; 26(12): 1878-1887, 2020 12.
Article in English | MEDLINE | ID: mdl-33020644

ABSTRACT

Autologous chimeric antigen receptor (CAR) T cell therapies targeting CD19 have high efficacy in large B cell lymphomas (LBCLs), but long-term remissions are observed in less than half of patients, and treatment-associated adverse events, such as immune effector cell-associated neurotoxicity syndrome (ICANS), are a clinical challenge. We performed single-cell RNA sequencing with capture-based cell identification on autologous axicabtagene ciloleucel (axi-cel) anti-CD19 CAR T cell infusion products to identify transcriptomic features associated with efficacy and toxicity in 24 patients with LBCL. Patients who achieved a complete response by positron emission tomography/computed tomography at their 3-month follow-up had three-fold higher frequencies of CD8 T cells expressing memory signatures than patients with partial response or progressive disease. Molecular response measured by cell-free DNA sequencing at day 7 after infusion was significantly associated with clinical response (P = 0.008), and a signature of CD8 T cell exhaustion was associated (q = 2.8 × 10-149) with a poor molecular response. Furthermore, a rare cell population with monocyte-like transcriptional features was associated (P = 0.0002) with high-grade ICANS. Our results suggest that heterogeneity in the cellular and molecular features of CAR T cell infusion products contributes to variation in efficacy and toxicity after axi-cel therapy in LBCL, and that day 7 molecular response might serve as an early predictor of CAR T cell efficacy.


Subject(s)
Cell- and Tissue-Based Therapy/adverse effects , Lymphoma, Large B-Cell, Diffuse/therapy , Receptors, Antigen, T-Cell/therapeutic use , Adult , Aged , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell-Free Nucleic Acids/blood , Female , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Neurotoxicity Syndromes/etiology , RNA-Seq , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis , Transcriptome/genetics
19.
Protein Cell ; 6(11): 814-24, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26487564

ABSTRACT

Ebolavirus can cause hemorrhagic fever in humans with a mortality rate of 50%-90%. Currently, no approved vaccines and antiviral therapies are available. Human TIM1 is considered as an attachment factor for EBOV, enhancing viral infection through interaction with PS located on the viral envelope. However, reasons underlying the preferable usage of hTIM-1, but not other PS binding receptors by filovirus, remain unknown. We firstly demonstrated a direct interaction between hTIM-1 and EBOV GP in vitro and determined the crystal structures of the Ig V domains of hTIM-1 and hTIM-4. The binding region in hTIM-1 to EBOV GP was mapped by chimeras and mutation assays, which were designed based on structural analysis. Pseudovirion infection assays performed using hTIM-1 and its homologs as well as point mutants verified the location of the GP binding site and the importance of EBOV GP-hTIM-1 interaction in EBOV cellular entry.


Subject(s)
Ebolavirus/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Flow Cytometry , Hepatitis A Virus Cellular Receptor 1 , Hepatitis A Virus Cellular Receptor 2 , Humans , Membrane Proteins/metabolism , Protein Binding , Surface Plasmon Resonance
20.
Protein Cell ; 5(9): 692-703, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24986489

ABSTRACT

Unlike the well-established picture for the entry of enveloped viruses, the mechanism of cellular entry of non-enveloped eukaryotic viruses remains largely mysterious. Picornaviruses are representative models for such viruses, and initiate this entry process by their functional receptors. Here we present the structural and functional studies of SCARB2, a functional receptor of the important human enterovirus 71 (EV71). SCARB2 is responsible for attachment as well as uncoating of EV71. Differences in the structures of SCARB2 under neutral and acidic conditions reveal that SCARB2 undergoes a pivotal pH-dependent conformational change which opens a lipid-transfer tunnel to mediate the expulsion of a hydrophobic pocket factor from the virion, a pre-requisite for uncoating. We have also identified the key residues essential for attachment to SCARB2, identifying the canyon region of EV71 as mediating the receptor interaction. Together these results provide a clear understanding of cellular attachment and initiation of uncoating for enteroviruses.


Subject(s)
Enterovirus A, Human/metabolism , Lysosomal Membrane Proteins/metabolism , Receptors, Scavenger/metabolism , Virion/metabolism , Virus Attachment , Acids/chemistry , Amino Acid Sequence , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/physiology , HEK293 Cells , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Molecular Docking Simulation , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Sequence Homology, Amino Acid , Sf9 Cells , Static Electricity , Virion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL