Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4419, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37479682

ABSTRACT

Variation in the antibody response has been linked to differential outcomes in disease, and suboptimal vaccine and therapeutic responsiveness, the determinants of which have not been fully elucidated. Countering models that presume antibodies are generated largely by stochastic processes, we demonstrate that polymorphisms within the immunoglobulin heavy chain locus (IGH) impact the naive and antigen-experienced antibody repertoire, indicating that genetics predisposes individuals to mount qualitatively and quantitatively different antibody responses. We pair recently developed long-read genomic sequencing methods with antibody repertoire profiling to comprehensively resolve IGH genetic variation, including novel structural variants, single nucleotide variants, and genes and alleles. We show that IGH germline variants determine the presence and frequency of antibody genes in the expressed repertoire, including those enriched in functional elements linked to V(D)J recombination, and overlapping disease-associated variants. These results illuminate the power of leveraging IGH genetics to better understand the regulation, function, and dynamics of the antibody response in disease.


Subject(s)
Genes, Immunoglobulin Heavy Chain , Genes, Immunoglobulin , Humans , Genes, Immunoglobulin Heavy Chain/genetics , Alleles , Germ-Line Mutation , Immunoglobulin Heavy Chains/genetics
2.
J Immunol ; 210(10): 1607-1619, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37027017

ABSTRACT

Current Adaptive Immune Receptor Repertoire sequencing (AIRR-seq) using short-read sequencing strategies resolve expressed Ab transcripts with limited resolution of the C region. In this article, we present the near-full-length AIRR-seq (FLAIRR-seq) method that uses targeted amplification by 5' RACE, combined with single-molecule, real-time sequencing to generate highly accurate (99.99%) human Ab H chain transcripts. FLAIRR-seq was benchmarked by comparing H chain V (IGHV), D (IGHD), and J (IGHJ) gene usage, complementarity-determining region 3 length, and somatic hypermutation to matched datasets generated with standard 5' RACE AIRR-seq using short-read sequencing and full-length isoform sequencing. Together, these data demonstrate robust FLAIRR-seq performance using RNA samples derived from PBMCs, purified B cells, and whole blood, which recapitulated results generated by commonly used methods, while additionally resolving H chain gene features not documented in IMGT at the time of submission. FLAIRR-seq data provide, for the first time, to our knowledge, simultaneous single-molecule characterization of IGHV, IGHD, IGHJ, and IGHC region genes and alleles, allele-resolved subisotype definition, and high-resolution identification of class switch recombination within a clonal lineage. In conjunction with genomic sequencing and genotyping of IGHC genes, FLAIRR-seq of the IgM and IgG repertoires from 10 individuals resulted in the identification of 32 unique IGHC alleles, 28 (87%) of which were previously uncharacterized. Together, these data demonstrate the capabilities of FLAIRR-seq to characterize IGHV, IGHD, IGHJ, and IGHC gene diversity for the most comprehensive view of bulk-expressed Ab repertoires to date.


Subject(s)
Complementarity Determining Regions , Humans , Complementarity Determining Regions/genetics , Base Sequence
3.
Nat Immunol ; 24(2): 239-254, 2023 02.
Article in English | MEDLINE | ID: mdl-36604547

ABSTRACT

Metastasis is the leading cause of cancer-related deaths and myeloid cells are critical in the metastatic microenvironment. Here, we explore the implications of reprogramming pre-metastatic niche myeloid cells by inducing trained immunity with whole beta-glucan particle (WGP). WGP-trained macrophages had increased responsiveness not only to lipopolysaccharide but also to tumor-derived factors. WGP in vivo treatment led to a trained immunity phenotype in lung interstitial macrophages, resulting in inhibition of tumor metastasis and survival prolongation in multiple mouse models of metastasis. WGP-induced trained immunity is mediated by the metabolite sphingosine-1-phosphate. Adoptive transfer of WGP-trained bone marrow-derived macrophages reduced tumor lung metastasis. Blockade of sphingosine-1-phosphate synthesis and mitochondrial fission abrogated WGP-induced trained immunity and its inhibition of lung metastases. WGP also induced trained immunity in human monocytes, resulting in antitumor activity. Our study identifies the metabolic sphingolipid-mitochondrial fission pathway for WGP-induced trained immunity and control over metastasis.


Subject(s)
Lung Neoplasms , beta-Glucans , Animals , Mice , Humans , Trained Immunity , Macrophages , Lysophospholipids/metabolism , Monocytes , Lung Neoplasms/pathology , beta-Glucans/metabolism , beta-Glucans/pharmacology , Tumor Microenvironment
4.
EBioMedicine ; 82: 104136, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35785620

ABSTRACT

BACKGROUND: Psoriasis recurrence is a clinically challenging issue. However, the underlying mechanisms haven't been fully understood. METHODS: RNAseq analysis from affected skin of psoriatic patients treated with topical glucocorticoid (GC) with different outcomes was performed. In addition, imiquimod (IMQ)-induced mouse psoriasis-like model was used to mimic GC treatment in human psoriasis patients. Skin tissues and draining and distant lymph nodes (LNs) were harvested for flow cytometry and histology analyses. FINDINGS: RNAseq analysis revealed that chemokine and chemokine receptor gene expression was decreased in post-treated skin compared to pre-treated samples but was subsequently increased in the recurred skin. In IMQ-induced mouse psoriasis-like model, we found that γδT17 cells were decreased in the skin upon topical GC treatment but surprisingly increased in the draining and distant LNs. This redistribution pattern lasted even two weeks post GC withdrawal. Upon IMQ re-challenge on the same site, mice previously treated with GC developed more severe skin inflammation. There were γδT17 cells migrated from LNs to the skin. This dynamic trafficking was dependent on CCR6 as this phenomenon was completely abrogated in CCR6-deficient mice. In addition, inhibition of lymphocyte egress prevented this heightened skin inflammation induced by IMQ rechallenge. INTERPRETATION: Redistribution of pathogenic γδT17 cells may be vital to prevent disease recurrence and this model of psoriasis-like dermatitis. FUNDING: This work was supported by National Natural Science Foundation of China 81830095/H1103, 81761128008/H10 (J.Z.) and the NIH R01AI128818 and the National Psoriasis Foundation (J.Y.).


Subject(s)
Dermatitis , Psoriasis , Animals , Dermatitis/metabolism , Dermatitis/pathology , Disease Models, Animal , Humans , Imiquimod/adverse effects , Inflammation/pathology , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Psoriasis/metabolism , Skin/pathology , T-Lymphocytes/metabolism
5.
Nat Commun ; 13(1): 759, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140221

ABSTRACT

Despite the remarkable success of immunotherapy in many types of cancer, pancreatic ductal adenocarcinoma has yet to benefit. Innate immune cells are critical to anti-tumor immunosurveillance and recent studies have revealed that these populations possess a form of memory, termed trained innate immunity, which occurs through transcriptomic, epigenetic, and metabolic reprograming. Here we demonstrate that yeast-derived particulate ß-glucan, an inducer of trained immunity, traffics to the pancreas, which causes a CCR2-dependent influx of monocytes/macrophages to the pancreas that display features of trained immunity. These cells can be activated upon exposure to tumor cells and tumor-derived factors, and show enhanced cytotoxicity against pancreatic tumor cells. In orthotopic models of pancreatic ductal adenocarcinoma, ß-glucan treated mice show significantly reduced tumor burden and prolonged survival, which is further enhanced when combined with immunotherapy. These findings characterize the dynamic mechanisms and localization of peripheral trained immunity and identify an application of trained immunity to cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Immunity , Pancreas/drug effects , Pancreatic Neoplasms/drug therapy , Animals , Bacteria , Female , Fungi , Immunity, Innate/immunology , Lectins, C-Type , Male , Mice , Myeloid Cells , Receptors, CCR2/genetics , beta-Glucans/immunology , Pancreatic Neoplasms
6.
Cell Metab ; 33(10): 2040-2058.e10, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34559989

ABSTRACT

One of the defining characteristics of a pre-metastatic niche, a fundamental requirement for primary tumor metastasis, is infiltration of immunosuppressive macrophages. How these macrophages acquire their phenotype remains largely unexplored. Here, we demonstrate that tumor-derived exosomes (TDEs) polarize macrophages toward an immunosuppressive phenotype characterized by increased PD-L1 expression through NF-kB-dependent, glycolytic-dominant metabolic reprogramming. TDE signaling through TLR2 and NF-κB leads to increased glucose uptake. TDEs also stimulate elevated NOS2, which inhibits mitochondrial oxidative phosphorylation resulting in increased conversion of pyruvate to lactate. Lactate feeds back on NF-κB, further increasing PD-L1. Analysis of metastasis-negative lymph nodes of non-small-cell lung cancer patients revealed that macrophage PD-L1 positively correlates with levels of GLUT-1 and vesicle release gene YKT6 from primary tumors. Collectively, our study provides a novel mechanism by which macrophages within a pre-metastatic niche acquire their immunosuppressive phenotype and identifies an important link among exosomes, metabolism, and metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Exosomes , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Exosomes/metabolism , Glycolysis , Humans , Lung Neoplasms/metabolism , Macrophages/metabolism , R-SNARE Proteins/metabolism , Tumor Microenvironment
7.
J Immunol ; 207(7): 1785-1797, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34470858

ABSTRACT

Our previous work has revealed the ability of CD11b to regulate BCR signaling and control autoimmune disease in mice. However, how CD11b regulates the immune response under normal conditions remains unknown. Through the use of a CD11b knockout model on a nonautoimmune background, we demonstrated that CD11b-deficient mice have an elevated Ag-specific humoral response on immunization. Deletion of CD11b resulted in elevated low-affinity and high-affinity IgG Ab and increases in Ag-specific germinal center B cells and plasma cells (PCs). Examination of BCR signaling in CD11b-deficient mice revealed defects in association of negative regulators pLyn and CD22 with the BCR, but increases in colocalizations between positive regulator pSyk and BCR after stimulation. Using a CD11b-reporter mouse model, we identified multiple novel CD11b-expressing B cell subsets that are dynamically altered during immunization. Subsequent experiments using a cell-specific CD11b deletion model revealed this effect to be B cell intrinsic and not altered by myeloid cell CD11b expression. Importantly, CD11b expression on PCs also impacts on BCR repertoire selection and diversity in autoimmunity. These studies describe a novel role for CD11b in regulation of the healthy humoral response and autoimmunity, and reveal previously unknown populations of CD11b-expressing B cell subsets, suggesting a complex function for CD11b in B cells during development and activation.


Subject(s)
B-Lymphocytes/immunology , CD11b Antigen/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , Autoimmunity , CD11b Antigen/genetics , Cells, Cultured , Humans , Immunity, Humoral , Immunization , Immunomodulation , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Receptors, Antigen, B-Cell/genetics , Signal Transduction , Syk Kinase/metabolism
8.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33986193

ABSTRACT

SARS coronavirus 2 (SARS-CoV-2) is a novel viral pathogen that causes a clinical disease called coronavirus disease 2019 (COVID-19). Although most COVID-19 cases are asymptomatic or involve mild upper respiratory tract symptoms, a significant number of patients develop severe or critical disease. Patients with severe COVID-19 commonly present with viral pneumonia that may progress to life-threatening acute respiratory distress syndrome (ARDS). Patients with COVID-19 are also predisposed to venous and arterial thromboses that are associated with a poorer prognosis. The present study identified the emergence of a low-density inflammatory neutrophil (LDN) population expressing intermediate levels of CD16 (CD16Int) in patients with COVID-19. These cells demonstrated proinflammatory gene signatures, activated platelets, spontaneously formed neutrophil extracellular traps, and enhanced phagocytic capacity and cytokine production. Strikingly, CD16Int neutrophils were also the major immune cells within the bronchoalveolar lavage fluid, exhibiting increased CXCR3 but loss of CD44 and CD38 expression. The percentage of circulating CD16Int LDNs was associated with D-dimer, ferritin, and systemic IL-6 and TNF-α levels and changed over time with altered disease status. Our data suggest that the CD16Int LDN subset contributes to COVID-19-associated coagulopathy, systemic inflammation, and ARDS. The frequency of that LDN subset in the circulation could serve as an adjunct clinical marker to monitor disease status and progression.


Subject(s)
Blood Coagulation Disorders/blood , Blood Coagulation Disorders/etiology , COVID-19/blood , COVID-19/complications , Neutrophils/immunology , SARS-CoV-2 , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Blood Coagulation Disorders/immunology , COVID-19/immunology , Cytokines/blood , Female , GPI-Linked Proteins/blood , Hospitalization , Humans , Inflammation Mediators/blood , Male , Middle Aged , Neutrophils/classification , Pandemics , Phagocytosis , Platelet Activation , Receptors, IgG/blood , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/immunology , Severity of Illness Index
9.
Cell Rep ; 33(11): 108503, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326773

ABSTRACT

Gut microbiota composition is associated with human and rodent Plasmodium infections, yet the mechanism by which gut microbiota affects the severity of malaria remains unknown. Humoral immunity is critical in mediating the clearance of Plasmodium blood stage infections, prompting the hypothesis that mice with gut microbiota-dependent decreases in parasite burden exhibit better germinal center (GC) responses. In support of this hypothesis, mice with a low parasite burden exhibit increases in GC B cell numbers and parasite-specific antibody titers, as well as better maintenance of GC structures and a more targeted, qualitatively different antibody response. This enhanced humoral immunity affects memory, as mice with a low parasite burden exhibit robust protection against challenge with a heterologous, lethal Plasmodium species. These results demonstrate that gut microbiota composition influences the biology of spleen GCs as well as the titer and repertoire of parasite-specific antibodies, identifying potential approaches to develop optimal treatments for malaria.


Subject(s)
Gastrointestinal Microbiome/immunology , Germinal Center/metabolism , Malaria/immunology , Animals , Humans , Mice
10.
J Clin Invest ; 130(4): 2081-2096, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31945018

ABSTRACT

Macrophages have been linked to tumor initiation, progression, metastasis, and treatment resistance. However, the transcriptional regulation of macrophages driving the protumor function remains elusive. Here, we demonstrate that the transcription factor c-Maf is a critical controller for immunosuppressive macrophage polarization and function in cancer. c-Maf controls many M2-related genes and has direct binding sites within a conserved noncoding sequence of the Csf-1r gene and promotes M2-like macrophage-mediated T cell suppression and tumor progression. c-Maf also serves as a metabolic checkpoint regulating the TCA cycle and UDP-GlcNAc biosynthesis, thus promoting M2-like macrophage polarization and activation. Additionally, c-Maf is highly expressed in tumor-associated macrophages (TAMs) and regulates TAM immunosuppressive function. Deletion of c-Maf specifically in myeloid cells results in reduced tumor burden with enhanced antitumor T cell immunity. Inhibition of c-Maf partly overcomes resistance to anti-PD-1 therapy in a subcutaneous LLC tumor model. Similarly, c-Maf is expressed in human M2 and tumor-infiltrating macrophages/monocytes as well as circulating monocytes of human non-small cell lung carcinoma (NSCLC) patients and critically regulates their immunosuppressive activity. The natural compound ß-glucan downregulates c-Maf expression on macrophages, leading to enhanced antitumor immunity in mice. These findings establish a paradigm for immunosuppressive macrophage polarization and transcriptional regulation by c-Maf and suggest that c-Maf is a potential target for effective tumor immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Immunity, Cellular , Lung Neoplasms/immunology , Macrophage Activation , Macrophages/immunology , Neoplasms, Experimental/immunology , Proto-Oncogene Proteins c-maf/immunology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Macrophages/pathology , Male , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-maf/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology
11.
J Invest Dermatol ; 139(1): 146-156, 2019 01.
Article in English | MEDLINE | ID: mdl-30120937

ABSTRACT

The IL-1 signaling pathway has been shown to play a critical role in the pathogenesis of chronic, autoinflammatory skin diseases such as psoriasis. However, the exact cellular and molecular mechanisms have not been fully understood. Here, we show that IL-1ß is significantly elevated in psoriatic lesional skin and imiquimod-treated mouse skin. In addition, IL-1R signaling appears to correlate with psoriasis disease progression and treatment response. IL-1 signaling in both dermal γδ T cells and other cells such as keratinocytes is essential to an IMQ-induced skin inflammation. IL-1ß induces dermal γδ T cell proliferation and IL-17 production in mice. In addition, IL-1ß stimulates keratinocytes to secrete chemokines that preferentially chemoattract peripheral CD27- CCR6+IL-17 capable of producing γδ T cells (γδT17). Further studies showed that endogenous IL-1ß secretion is regulated by skin commensals to maintain dermal γδT17 homeostasis in mice. Mouse skin associated with Corynebacterium species, bacteria enriched in human psoriatic lesional skin, has increased IL-1ß and dermal γδT17 cell expansion. Thus, the IL-1ß-IL-1R signaling pathway may contribute to skin inflammation and psoriasis pathogenesis via the direct regulation of dermal IL-17-producing cells and stimulation of keratinocytes for amplifying inflammatory cascade.


Subject(s)
Gene Expression Regulation , Inflammation/genetics , Interleukin-1beta/genetics , Keratinocytes/metabolism , Psoriasis/genetics , RNA/genetics , Receptors, Interleukin-1 Type I/genetics , Animals , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/blood , Keratinocytes/pathology , Mice , Mice, Knockout , Polymerase Chain Reaction , Psoriasis/metabolism , Psoriasis/pathology , Receptors, Interleukin-1 Type I/biosynthesis
12.
PLoS One ; 13(8): e0201813, 2018.
Article in English | MEDLINE | ID: mdl-30089167

ABSTRACT

Breast cancer (BC) is increasing in incidence and resistance to treatment worldwide. The challenges in limited therapeutic options and poor survival outcomes in BC subtypes persist because of its molecular heterogeneity and resistance to standard endocrine therapy. Recently, high throughput RNA sequencing (RNA-seq) has been used to identify biomarkers of disease progression and signaling pathways that could be amenable to specific therapies according to the BC subtype. However, there is no single generally accepted pipeline for the analysis of RNA-seq data in biomarker discovery due, in part, to the needs of simultaneously satisfying constraints of sensitivity and specificity. We proposed a combined approach using gene-wise normalization, UQ-pgQ2, followed by a Wald test from DESeq2. Our approach improved the analysis based on within-group comparisons in terms of the specificity when applied to publicly available RNA-seq BC datasets. In terms of identifying differentially expressed genes (DEGs), we combined an optimized log2 fold change cutoff with a nominal false discovery rate of 0.05 to further minimize false positives. Using this method in the analysis of two GEO BC datasets, we identified 797 DEGs uniquely expressed in triple negative BC (TNBC) and significantly associated with T cell and immune-related signaling, contributing to the immunotherapeutic efficacy in TNBC patients. In contrast, we identified 1403 DEGs uniquely expressed in estrogen positive and HER2 negative BC (ER+HER2-BC) and significantly associated with eicosanoid, notching and FAK signaling while a common set of genes was associated with cellular growth and proliferation. Thus, our approach to control for false positives identified two distinct gene expression profiles associated with these two subtypes of BC which are distinguishable by their molecular and functional attributes.


Subject(s)
Breast Neoplasms/metabolism , Sequence Analysis, RNA/methods , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/classification , Breast Neoplasms/genetics , Computational Biology , Gene Expression Regulation, Neoplastic , Humans , RNA, Messenger/metabolism
13.
Immunohorizons ; 2(11): 349-362, 2018 12 05.
Article in English | MEDLINE | ID: mdl-31026806

ABSTRACT

A mutation in STAT3 has been linked to the incidence of autosomal dominant hyper IgE syndrome, a disease characterized by elevated serum IgE Ab. However, how this genetic mutation leads to the phenotype has not been fully understood. We investigated the specific role of STAT3 in the germinal center (GC) B cells and plasma cells for IgE class switching. Through the use of STAT3 conditional knockout (cKO) mice in a Th2-type immunization model, we demonstrated that CD2-Cre-driven STAT3 cKO mice showed elevated IgE and decreased IgG1 in the serum and a reduction in GC formation. Within the GC, IgG1 + GC B cells were decreased, whereas IgE+ GC B cells were more prevalent. Additionally, these mice exhibited reduced IgG1 and elevated IgE populations of Ab-producing plasma cells. Subsequent experiments using a CD19-Cre cKO mouse established this effect to be B cell-intrinsic. Transcription factors critical for GC and plasma cell differentiation, including Bcl-6 and Aicda, were shown to function as downstream signals of STAT3 regulation. Chromatin immunoprecipitation sequencing analysis revealed that many genes, including Bcl3 and Crtc2, were among the direct STAT3 regulated targets. Mice with STAT3 deficiency in B cells also demonstrated an increase in lung inflammation when used in an asthma-like disease model. This model suggests a negative role for STAT3 in regulating class switching of the GC B cells from the IgG1 to the IgE producing state, which may serve as a therapeutic target for treatment of autosomal dominant hyper IgE syndrome and other immune disorders.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunoglobulin Class Switching , Immunoglobulin E/immunology , Immunoglobulin G/immunology , STAT3 Transcription Factor/immunology , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , B-Lymphocytes/cytology , Cell Differentiation/immunology , Disease Models, Animal , Germinal Center/metabolism , Immunoglobulin E/genetics , Immunoglobulin G/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/cytology , Plasma Cells/immunology , Pneumonia/immunology , Primary Cell Culture , STAT3 Transcription Factor/genetics , Spleen/immunology
14.
Phys Rev Lett ; 114(9): 093002, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25793810

ABSTRACT

As an alternative to state-of-the-art laser frequency stabilization using ultrastable cavities, it has been proposed to exploit the nonlinear effects from coupling of atoms with a narrow transition to an optical cavity. Here, we have constructed such a system and observed nonlinear phase shifts of a narrow optical line by a strong coupling of a sample of strontium-88 atoms to an optical cavity. The sample temperature of a few mK provides a domain where the Doppler energy scale is several orders of magnitude larger than the narrow linewidth of the optical transition. This makes the system sensitive to velocity dependent multiphoton scattering events (Dopplerons) that affect the cavity field transmission and phase. By varying the number of atoms and the intracavity power, we systematically study this nonlinear phase signature which displays roughly the same features as for much lower temperature samples. This demonstration in a relatively simple system opens new possibilities for alternative routes to laser stabilization at the sub-100 mHz level and superradiant laser sources involving narrow-line atoms. The understanding of relevant motional effects obtained here has direct implications for other atomic clocks when used in relation to ultranarrow clock transitions.

SELECTION OF CITATIONS
SEARCH DETAIL
...